Language selection

Search

Patent 2386190 Summary

Third-party information liability

Some of the information on this Web page has been provided by external sources. The Government of Canada is not responsible for the accuracy, reliability or currency of the information supplied by external sources. Users wishing to rely upon this information should consult directly with the source of the information. Content provided by external sources is not subject to official languages, privacy and accessibility requirements.

Claims and Abstract availability

Any discrepancies in the text and image of the Claims and Abstract are due to differing posting times. Text of the Claims and Abstract are posted:

  • At the time the application is open to public inspection;
  • At the time of issue of the patent (grant).
(12) Patent: (11) CA 2386190
(54) English Title: REGULATORS OF THE HEDGEHOG PATHWAY, COMPOSITIONS AND USES RELATED THERETO
(54) French Title: REGULATEURS DU SIGNAL HEDGEHOG, COMPOSITIONS ET UTILISATIONS DESDITS REGULATEURS
Status: Term Expired - Post Grant Beyond Limit
Bibliographic Data
(51) International Patent Classification (IPC):
  • C07J 69/00 (2006.01)
  • A61K 31/58 (2006.01)
  • A61P 35/00 (2006.01)
(72) Inventors :
  • BEACHY, PHILIP A. (United States of America)
  • CHEN, JAMES K. (United States of America)
  • TAIPALE, ANSSI J. (United States of America)
(73) Owners :
  • JOHNS HOPKINS UNIVERSITY SCHOOL OF MEDICINE
(71) Applicants :
  • JOHNS HOPKINS UNIVERSITY SCHOOL OF MEDICINE (United States of America)
(74) Agent: BORDEN LADNER GERVAIS LLP
(74) Associate agent:
(45) Issued: 2009-04-14
(86) PCT Filing Date: 2000-10-13
(87) Open to Public Inspection: 2001-04-19
Examination requested: 2004-05-04
Availability of licence: N/A
Dedicated to the Public: N/A
(25) Language of filing: English

Patent Cooperation Treaty (PCT): Yes
(86) PCT Filing Number: PCT/US2000/028479
(87) International Publication Number: US2000028479
(85) National Entry: 2002-04-11

(30) Application Priority Data:
Application No. Country/Territory Date
60/159,215 (United States of America) 1999-10-13
60/229,273 (United States of America) 2000-08-30

Abstracts

English Abstract


The present invention makes available, inter alia, methods and reagents for
modulating smoothened-dependent pathway
activation. In certain embodiments, the subject methods can be used to
counteract the phenotypic effects of unwanted activation
of a hedgehog pathway, such as resulting from hedgehog gain-of-function, ptc
loss-of-function or smoothened gain-of-function mutations.


French Abstract

La présente invention concerne, entre autres, des méthodes et des réactifs pouvant moduler l'activité du signal Smoothened. Dans certaines formes de réalisation, lesdites méthodes peuvent être utilisées pour contrecarrer les effets phénotypiques d'une activité non désirée d'un signal Hedgehog, telle que résultant de mutations par gain-de-fonction Hedgehog, par perte-de-fonction ptc, ou par gain-de-fonction Smoothened.

Claims

Note: Claims are shown in the official language in which they were submitted.


134
CLAIMS:
1. A compound represented in the general formula (IV) or unsaturated forms
thereof:
<IMG>
wherein, as valence and stability permit,
R2 and R3, independently for each occurrence, represent one or more
substitutions to
the ring to which each is attached, wherein the substitutions comprise 0 to 30
carbon atoms and are hydrogen, halogens, alkyls, alkenyls, alkynyls, aryls,
hydroxyl, =O, =S, alkoxyl, silyloxy, amino, nitro, thiol, amines, imines,
amides, phosphoryls, phosphonates, phosphines, carbonyls, carboxyls,
carboxamides, anhydrides, silyls, ethers, thioethers, alkylsulfonyls,
arylsulfonyls, selenoethers, ketones, aldehydes, esters, sugar or -(CH2)m-R8;
R4 and R5, independently for each occurrence, are absent or represent one or
more
substitutions to the ring to which each is attached, wherein the substitutions
comprise 0 to 30 carbon atoms and are hydrogen, halogens, alkyls, alkenyls,
alkynyls, aryls, hydroxyl, =O, =S, alkoxyl, silyloxy, amino, nitro, thiol,
amines, imines, amides, phosphoryls, phosphonates, phosphines, carbonyls,
carboxyls, carboxamides, anhydrides, silyls, ethers, thioethers,
alkylsulfonyls,
arylsulfonyls, selenoethers, ketones, aldehydes, esters, sugar, carbamate,
carbonate, or -(CH2)m-R8;
R6 is absent or represents, independently, a substituent comprising 0 to 30
carbon
atoms which is halogen, alkyl, alkenyl, alkynyl, aryl, hydroxyl, =O, =S,
alkoxyl, silyloxy, amino, nitro, thiol, amine, imine, amide, phosphoryl,
phosphonate, phosphine, carbonyl, carboxyl, carboxamide, anhydride, silyl,

135
ether, thioether, alkylsulfonyl, arylsulfonyl, selenoether, ketone, aldehyde,
ester, or -(CH2)m-R8;
R8 represents a substituent comprising 0 to 30 carbon atoms comprising an
aryl, a
cycloalkyl, a cycloalkenyl, a heterocycle, or a polycycle;
R9 independently for each occurrence, represents one or more substitutions to
the ring
to which it is attached, wherein the substitutions comprise 0 to 30 carbon
atoms and are halogen, alkyl, alkenyl, alkynyl, aryl, hydroxyl, =O, =S,
alkoxyl, silyloxy, amino, nitro, thiol, amine, imine, amide, phosphoryl,
phosphonate, phosphine, carbonyl, carboxyl, carboxamide, anhydride, silyl,
ether, thioether, alkylsulfonyl, arylsulfonyl, selenoether, ketone, aldehyde,
ester, or -(CH2)m-R8;
R22 is absent or represents a substituent comprising 0 to 30 carbon atoms
comprising
an alkyl, an alkoxyl or -OH; and
m is an integer in the range 0 to 8 inclusive,
wherein at least one occurrence of R9 is bound to N, thereby forming a
tertiary amine
with an extraannular substituent, and this occurrence of R9 is an alkyl
substituted with an acylamino;
wherein alkyl includes saturated aliphatic groups, straight-chain alkyl
groups,
branched-chain alkyl groups, cycloalkyl (alicyclic) groups, alkyl-substituted
cycloalkyl groups, and cycloalkyl-substituted alkyl groups, wherein alkyl
includes substituted and unsubstituted alkyl groups;
wherein amine and amino includes both substituted and unsubstituted amines and
aminos; and
wherein heterocycle refers to 3- to 10-membered ring structures whose ring
structures
include one to four heteroatoms;
or a pharmaceutically acceptable salt thereof.
2. The compound of claim 1, wherein:
R2 represents =O, sugar, carbamate, ester, carbonate, or alkoxy;
R3, for each occurrence, is an -OH, alkyl, -O-alkyl, -C(O)-alkyl, or -C(O)-R8;
R4, for each occurrence, is absent, or represents -OH, =O, alkyl, -O-alkyl, -
C(O)-
alkyl, or -C(O)-R8; and

136
R5, for each occurrence, is absent, or represents -OH, =O, or alkyl.
3. A compound represented in the general formula (V) or unsaturated forms
thereof:
<IMG>
wherein, as valence and stability permit,
R2 and R3, independently for each occurrence, represent one or more
substitutions to
the ring to which each is attached, wherein the substitutions comprise 0 to 30
carbon atoms and are hydrogen, halogens, alkyls, alkenyls, alkynyls, aryls,
hydroxyl, =O, =S, alkoxyl, silyloxy, amino, nitro, thiol, amines, imines,
amides, phosphoryls, phosphonates, phosphines, carbonyls, carboxyls,
carboxamides, anhydrides, silyls, ethers, thioethers, alkylsulfonyls,
arylsulfonyls, selenoethers, ketones, aldehydes, esters, sugar or -(CH2)m-R8;
R4 is absent or represents one or more substitutions to the ring to which each
is
attached, wherein the substitutions comprise 0 to 30 carbon atoms and are
hydrogen, halogens, alkyls, alkenyls, alkynyls, aryls, hydroxyl, =O, =S,
alkoxyl, silyloxy, amino, nitro, thiol, amines, imines, amides, phosphoryls,
phosphonates, phosphines, carbonyls, carboxyls, carboxamides, anhydrides,
silyls, ethers, thioethers, alkylsulfonyls, arylsulfonyls, selenoethers,
ketones,
aldehydes, esters, sugar, carbamate, carbonate, or -(CH2)m-R8;
R6 is absent or represents a substituent comprising 0 to 30 carbon atoms and
is
halogen, alkyl, alkenyl, alkynyl, aryl, hydroxyl, =O, =S, alkoxyl, silyloxy,
amino, nitro, thiol, amine, imine, amide, phosphoryl, phosphonate, phosphine,
carbonyl, carboxyl, carboxamide, anhydride, silyl, ether, thioether,

137
alkylsulfonyl, arylsulfonyl, selenoether, ketone, aldehyde, ester, or -(CH2)m-
R8;
R8 represents a substituent comprising 0 to 30 carbon atoms comprising an
aryl, a
cycloalkyl, a cycloalkenyl, a heterocycle, or a polycycle;
R9 independently for each occurrence, represents one or more substitutions to
the ring
to which it is attached, wherein the substitutions comprise 0 to 30 carbon
atoms and are halogen, alkyl, alkenyl, alkynyl, aryl, hydroxyl, =O, =S,
alkoxyl, silyloxy, amino, nitro, thiol, amine, imine, amide, phosphoryl,
phosphonate, phosphine, carbonyl, carboxyl, carboxamide, anhydride, silyl,
ether, thioether, alkylsulfonyl, arylsulfonyl, selenoether, ketone, aldehyde,
ester, or -(CH2)m-R8; and
m is an integer in the range 0 to 8 inclusive,
wherein at least one occurrence of R9 is an extraannular substituent attached
to N,
thereby forming a tertiary amine, and this occurrence of R9 is an alkyl
substituted with an acylamino;
wherein alkyl includes saturated aliphatic groups, straight-chain alkyl
groups,
branched-chain alkyl groups, cycloalkyl (alicyclic) groups, alkyl-substituted
cycloalkyl groups, and cycloalkyl-substituted alkyl groups, wherein alkyl
includes substituted and unsubstituted alkyl groups;
wherein amine and amino includes both substituted and unsubstituted amines and
aminos; and
wherein heterocycle refers to 3- to 10-membered ring structures whose ring
structures
include one to four heteroatoms;
or a pharmaceutically acceptable salt thereof.
4. The compound of claim 3, wherein:
R2 represents =O, sugar, carbamate, ester, carbonate, or alkoxy;
R3, for each occurrence, is an -OH, alkyl, -O-alkyl, -C(O)-alkyl, or -C(O)-R8;
and
R4, for each occurrence, is absent, or represents -OH, =O, alkyl, -O-alkyl, -
C(O)-
alkyl, or -C(O) R8.

138
5. The compound of any one of claims 1-4, wherein the extraannular substituent
is hydrophobic.
6. The compound of any one of claims 1-4, wherein the extraannular substituent
comprises an aryl, heteroaryl, carbocyclyl, heterocyclyl, or polycyclyl group.
7. The compound of any one of claims 1-4, wherein the extraannular substituent
comprises a polycyclyl group which is biotin, a zwitterionic complex of boron,
or a
steroidal polycycle.
8. The compound of any one of claims 1-4, wherein the extraannular substituent
comprises an alkyl substituted with acylamino.
9. The compound of claim 5, wherein the hydrophobic substituent comprises a
combination of alkyl, amido, acylamino, ketone, ester, ether, halogen,
alkenyl,
alkynyl, aryl, aralkyl, urea, comprising between 5 and 40 non-hydrogen atoms.
10. Use of a compound of any one of claims 1-9 in the manufacture of a
medicament for treating basal cell carcinoma.
11. The use of claim 10, wherein the medicament is in locally administrable
form.
12. Use of a compound of any one of claims 1-9 in the manufacture of a
medicament for regulating differentiation or proliferation of a cell.
13. Use of a compound of any one of claims 1-9 in the manufacture of a
medicament for controlling the growth or development of pancreatic tissue.
14. Use of a compound of any one of claims 1-9 in the manufacture of a
medicament for treating medulloblastoma.
15. The use of claim 14, wherein the medicament is in locally administrable
form.

139
16. Use of a compound of any one of claims 1-9 in the manufacture of a
medicament for treating a hyperproliferative disorder.
17. The use of claim 16, wherein the medicament is in topically administrable
form.
18. The use of claim 16, wherein the medicament is in locally administrable
form.
19. A pharmaceutical preparation comprising a compound of any one of claims 1-
9 and a pharmaceutically acceptable excipient.
20. Use of a compound of any one of claims 1-9 in the manufacture of a
medicament for inhibiting hedgehog signaling or counteracting a ptc loss-of-
function
phenotype or a smoothened gain-of-function phenotype in a cell.
21. The use of claim 20, wherein the compound inhibits ptc loss-of-function or
smoothened gain-of-function mediated signal transduction with an ED50 of 1 mM
or
less.
22. The use of claim 20, wherein the compound inhibits ptc loss-of-function or
smoothened gain-of-function mediated signal transduction with an ED50 of 1
µM or
less.
23. The use of claim 20, wherein the compound inhibits ptc loss-of-function or
smoothened gain-of-function mediated signal transduction with an ED50 of 1 nM
or
less.
24. The use of claim 20, wherein the medicament is for therapeutic or cosmetic
application.
25. The use of claim 24, wherein the therapeutic or cosmetic application is
regulation of neural tissues, bone and cartilage formation and repair,
regulation of

140
spermatogenesis, regulation of smooth muscle, regulation of lung, liver and
gut,
regulation of hematopoietic function, or regulation of skin or hair growth.

Description

Note: Descriptions are shown in the official language in which they were submitted.


CA 02386190 2006-08-30
-1-
REGULATORS OF THE HEDGEHOG PATHWAY,
COMPOSITIONS AND USES RELATED THERETO
Background of the Invention
Pattern formation is the activity by which embryonic cells fonn ordered
spatial
arrangements of differentiated tissues. The physical complexity of higher
organisms
arises during embryogenesis through the interplay of cell-intrinsic lineage
and cell-
extrinsic signaling. Inductive interactions are essential to embryonic
patteming in
vertebrate development from the earliest establishment of the body plan, to
the
patteming of the organ systems, to the generation of diverse cell types during
tissue
differentiation (Davidson, E., (1990) Development 108: 365-389; Gurdon, J. B.,
(1992)
Cell 68: 185-199; Jessell, T. M. et al., (1992) Cell 68: 257-270). The effects
of
developmental cell interactions are varied. Typically, responding cells are
diverted from
one route of cell differentiation to another by inducing cells that differ
from both the
uninduced and induced states of the responding cells (inductions). Sometimes
cells
induce their neighbors to differentiate like themselves (homeogenetic
induction); in
other cases a cell inhibits its neighbors from differentiating like itself.
Cell interactions
in early development may be sequential, such that an initial induction between
two cell
types leads to a progressive amplification of diversity. Moreover, inductive
interactions
occur not only in embryos, but in adult cells as well, and can act to
establish and
maintain morphogenetic patterns as well as induce differentiation (J.B. Gurdon
(1992)
Ce1168:185-199).
Members of the Hedgehog family of signaling molecules mediate many
important short- and long-range patterning processes during invertebrate and
vertebrate
development. In the fly, a single hedgehog gene regulates segmental and
imaginal disc
patterning. In contrast, in vertebrates, a hedgehog gene family is involved in
the control

CA 02386190 2002-04-11
WO 01/27135 PCT/US00/28479
-2-
of left-right asymmetry, polarity in the CNS, somites and limb, organogenesis,
chondrogenesis and spermatogenesis.
The first hedgehog gene was identified by a genetic screen in the fruitfly
Drosophila melanogaster (Nusslein-Volhard, C. and Wieschaus, E. (1980) Nature
287,
795-801). This screen identified a number of mutations affecting embryonic and
larval
development. In 1992 and 1993, the molecular nature of the Drosophila hedgehog
(hh)
gene was reported (C.F., Lee et al. (1992) Cell 71, 33-50), and since then,
several
hedgehog homologues have been isolated from various vertebrate species. While
only
one hedgehog gene has been found in Drosophila and other invertebrates,
multiple
Hedgehog genes are present in vertebrates.
The vertebrate family of hedgehog genes includes at least four members, e.g.,
paralogs of the single drosophila hedgehog gene. Exemplary hedgehog genes and
proteins are described in PCT publications WO 95/18856 and WO 96/17924. Three
of
these members, herein referred to as Desert hedgehog (Dhh), Sonic hedgehog
(Shh) and
Indian hedgehog (Ihh), apparently exist in all vertebrates, including fish,
birds, and
mammals. A fourth member, herein referred to as tiggy-winkle hedgehog (Twhh),
appears specific to fish. Desert hedgehog (Dhh) is expressed principally in
the testes,
both in mouse embryonic development and in the adult rodent and human; Indian
hedgehog (Ihh) is involved in bone development during embryogenesis and in
bone
formation in the adult; and, Shh, which as described above, is primarily
involved in
morphogenic and neuroinductive activities. Given the critical inductive roles
of
hedgehog polypeptides in the development and maintenance of vertebrate organs,
the
identification of hedghog interacting proteins is of paramount significance in
both
clinical and research contexts.
The various Hedgehog proteins consist of a signal peptide, a highly conserved
N-
terminal region, and a more divergent C-terminal domain. In addition to signal
sequence
cleavage in the secretory pathway (Lee, J.J. et al. (1992) Ce1171:33-50;
Tabata, T. et al.
(1992) Genes Dev. 2635-2645; Chang, D.E. et al. (1994) Development 120:3339-
3353),
Hedgehog precursor proteins undergo an internal autoproteolytic cleavage which
depends on conserved sequences in the C-terminal portion (Lee et al. (1994)
Science
266:1528-1537; Porter et al. (1995) Nature 374:363-366). This autocleavage
leads to a

CA 02386190 2002-04-11
WO 01/27135 PCT/US00/28479
-3-
19 kD N-terminal peptide and a C-terminal peptide of 26-28 kD (Lee et al.
(1992) supra;
Chang et al. (1994) supra; Lee et al. (1994) supra; Bumcrot, D.A., et al.
(1995) Mol.
Cell. Biol. 15:2294-2303; Porter et al. (1995) supra; Ekker, S.C. et al.
(1995) Curr. Biol.
5:944-955; Lai, C.J. et al. (1995) Development 121:2349-2360). The N-terminal
peptide
stays tightly associated with the surface of cells in which it was
synthesized, while the
C-terminal peptide is freely diffusible both in vitro and in vivo (Porter et
al. (1995)
Nature 374:363; Lee et al. (1994) supra; Bumcrot et al. (1995) su ra= Marti,
E. et al.
(1995) Development 121:2537-2547; Roelink, H. et al. (1995) Cell 81:445-455).
Interestingly, cell surface retention of the N-terminal peptide is dependent
on
autocleavage, as a truncated form of HH encoded by an RNA which terminates
precisely
at the normal position of internal cleavage is diffusible in vitro (Porter et
al. (1995)
supra) and in vivo (Porter, J.A. et al. (1996) Cell 86, 21-34). Biochemical
studies have
shown that the autoproteolytic cleavage of the HH precursor protein proceeds
through an
internal thioester intermediate which subsequently is cleaved in a
nucleophilic
substitution. It is likely that the nucleophile is a small lipophilic molecule
which
becomes covalently bound to the C-terminal end of the N-peptide (Porter et al.
(1996)
supra), tethering it to the cell surface. The biological implications are
profound. As a
result of the tethering, a high local concentration of N-terminal Hedgehog
peptide is
generated on the surface of the Hedgehog producing cells. It is this N-
terminal peptide
which is both necessary and sufficient for short- and long-range Hedgehog
signaling
activities in Drosophila and vertebrates (Porter et al. (1995) suQra; Ekker et
al. (1995)
supra; Lai et al. (1995) su ra; Roelink, H. et al. (1995) Cell 81:445-455;
Porter et al.
(1996) supra; Fietz, M.J. et al. (1995) Curr. Biol. 5:643-651; Fan, C.-M. et
al. (1995)
Cell 81:457-465; Marti, E., et al. (1995) Nature 375:322-325; Lopez-Martinez
et al.
(1995) Curr. Biol 5:791-795; Ekker, S.C. et al. (1995) Development 121:2337-
2347;
Forbes, A.J. et al.(1996) Development 122:1125-1135).
HH has been implicated in short- and long-range patterning processes at
various
sites during Drosophila development. In the establishment of segment polarity
in early
embryos, it has short-range effects which appear to be directly mediated,
while in the
patterning of the imaginal discs, it induces long range effects via the
induction of
secondary signals.

CA 02386190 2002-04-11
WO 01/27135 PCT/US00/28479
-4-
In vertebrates, several hedgehog genes have been cloned in the past few years.
Of
these genes, Shh has received most of the experimental attention, as it is
expressed in
different organizing centers which are the sources of signals that pattern
neighboring
tissues. Recent evidence indicates that Shh is involved in these interactions.
The expression of Shh starts shortly after the onset of gastrulation in the
presumptive midline mesoderm, the node in the mouse (Chang et al. (1994)
supra;
Echelard, Y. et al. (1993) Cell 75:1417-1430), the rat (Roelink, H. et al.
(1994) Cell
76:761-775) and the chick (Riddle, R.D. et al. (1993) Ce1175:1401-1416), and
the shield
in the zebrafish (Ekker et al. (1995) supra; Krauss, S. et al.(1993)
Ce1175:1431-1444). In
chick embyros, the Shh expression pattern in the node develops a left-right
asymmetry,
which appears to be responsible for the left-right situs of the heart (Levin,
M. et al.
(1995) Cell 82:803-814).
In the CNS, Shh from the notochord and the floorplate appears to induce
ventral
cell fates. When ectopically expressed, Shh leads to a ventralization of large
regions of
the mid- and hindbrain in mouse (Echelard et al. (1993) supra; Goodrich, L.V.
et al.
(1996) Genes Dev. 10:301-312), Xenopus (Roelink, H. et al. (1994) supra; Ruiz
i
Altaba, A. et al. (1995) Mol. Cell. Neurosci. 6:106-121), and zebrafish (Ekker
et al.
(1995) supra; Krauss et al. (1993) supra; Hammerschmidt, M., et al. (1996)
Genes Dev.
10:647-658). In explants of intermediate neuroectoderm at spinal cord levels,
Shh
protein induces floorplate and motor neuron development with distinct
concentration
thresholds, floor plate at high and motor neurons at lower concentrations
(Roelink et al.
(1995) supra; Marti et al. (1995) supra; Tanabe, Y. et al. (1995) Curr. Biol.
5:651-658).
Moreover, antibody blocking suggests that Shh produced by the notochord is
required
for notochord-mediated induction of motor neuron fates (Marti et al. (1995)
supra).
Thus, high concentration of Shh on the surface of Shh-producing midline cells
appears to
account for the contact-mediated induction of floorplate observed in vitro
(Placzek, M.
et al. (1993) Development 117:205-218), and the midline positioning of the
floorplate
immediately above the notochord in vivo. Lower concentrations of Shh released
from the
notochord and the floorplate presumably induce motor neurons at more distant
ventrolateral regions in a process that has been shown to be contact-
independent in vitro
(Yamada, T. et al. (1993) Cell 73:673-686). In explants taken at midbrain and
forebrain

CA 02386190 2002-04-11
WO 01/27135 PCT/US00/28479
-5-
levels, Shh also induces the appropriate ventrolateral neuronal cell types,
dopaminergic
(Heynes, M. et al. (1995) Neuron 15:35-44; Wang, M.Z. et al. (1995) Nature
Med.
1:1184-1188) and cholinergic (Ericson, J. et al. (1995) Cell 81:747-756)
precursors,
respectively, indicating that Shh is a common inducer of ventral specification
over the
entire length of the CNS. These observations raise a question as to how the
differential
response to Shh is regulated at particular anteroposterior positions.
Shh from the midline also patterns the paraxial regions of the vertebrate
embryo,
the somites in the trunk (Fan et al. (1995) supra) and the head mesenchyme
rostral of the
somites (Hammerschmidt et al. (1996) supra). In chick and mouse paraxial
mesoderm
explants, Shh promotes the expression of sclerotome specific markers like Paxl
and
Twist, at the expense of the dermamyotomal marker Pax3. Moreover, filter
barrier
experiments suggest that Shh mediates the induction of the sclerotome directly
rather
than by activation of a secondary signaling mechanism (Fan, C.-M. and Tessier-
Lavigne,
M. (1994) Ce1179, 1175-1186).
Shh also induces myotomal gene expression (Hammerschmidt et al. (1996)
supra; Johnson, R.L. et al. (1994) Cell 79:1165-1173; Miinsterberg, A.E. et
al. (1995)
Genes Dev. 9:2911-2922; Weinberg, E.S. et al. (1996) Development 122:271-280),
although recent experiments indicate that members of the WNT family,
vertebrate
homologues of Drosophila wingless, are required in concert (Munsterberg et al.
(1995)
sunra). Puzzlingly, myotomal induction in chicks requires higher Shh
concentrations
than the induction of sclerotomal markers (Munsterberg et al. (1995) supra),
although
the sclerotome originates from somitic cells positioned much closer to the
notochord.
Similar results were obtained in the zebrafish, where high concentrations of
Hedgehog
induce myotomal and repress sclerotomal marker gene expression (Hammerschmidt
et
al. (1996) supra). In contrast to amniotes, however, these observations are
consistent
with the architecture of the fish embryo, as here, the myotome is the
predominant and
more axial component of the somites. Thus, modulation of Shh signaling and the
acquisition of new signaling factors may have modified the somite structure
during
vertebrate evolution.
In the vertebrate limb buds, a subset of posterior mesenchymal cells, the
"Zone
of polarizing activity" (ZPA), regulates anteroposterior digit identity
(reviewed in Honig,

CA 02386190 2002-04-11
WO 01/27135 PCT/US00/28479
-6-
L.S. (1981) Nature 291:72-73). Ectopic expression of Shh or application of
beads soaked
in Shh peptide mimics the effect of anterior ZPA grafts, generating a mirror
image
duplication of digits (Chang et al. (1994) supra; Lopez-Martinez et al. (1995)
supra;
Riddle et al. (1993) supra) (Fig. 2g). Thus, digit identity appears to depend
primarily on
Shh concentration, although it is possible that other signals may relay this
information
over the substantial distances that appear to be required for AP patterning
(100-150 m).
Similar to the interaction of HH and DPP in the Drosophila imaginal discs, Shh
in the
vertebrate limb bud activates the expression of Bmp2 (Francis, P.H. et al.
(1994)
Development 120:209-218), a dpp homologue. However, unlike DPP in Drosophila,
Bmp2 fails to mimic the polarizing effect of Shh upon ectopic application in
the chick
limb bud (Francis et al. (1994) su ra). In addition to anteroposterior
patterning, Shh also
appears to be involved in the regulation of the proximodistal outgrowth of the
limbs by
inducing the synthesis of the fibroblast growth factor FGF4 in the posterior
apical
ectodermal ridge (Laufer, E. et al. (1994) Cell 79:993-1003; Niswander, L. et
al.(1994)
Nature 371:609-612).
The close relationship between Hedgehog proteins and BMPs is likely to have
been conserved at many, but probably not all sites of vertebrate Hedgehog
expression.
For example, in the chick hindgut, Shh has been shown to induce the expression
of
Bmp4, another vertebrate dpp homologue (Roberts, D.J. et al. (1995)
Development
121:3163-3174). Furthermore, Shh and Bmp2, 4, or 6 show a striking correlation
in their
expression in epithelial and mesenchymal cells of the stomach, the urogential
system,
the lung, the tooth buds and the hair follicles (Bitgood, M.J. and McMahon,
A.P. (1995)
Dev. Biol. 172:126-138). Further, Ihh, one of the two other mouse Hedgehog
genes, is
expressed adjacent to Bmp expressing cells in the gut and developing cartilage
(Bitgood
and McMahon (1995) supra).
A major function of hedgehog in the Drosophila embryo is the maintenance of
wg transcription at the boundary of each segmental unit (Hidalgo and Ingham,
(1990)
Development 110:291-302); from here, Wg protein diffuses across the segment to
specify the character of the ectodermal cells that secrete the larval cuticle
(Lawrence et
al., (1996) Development 122:4095-4103). Like hh, mutations in three other
segment

CA 02386190 2002-04-11
WO 01/27135 PCT/US00/28479
-7-
polarity genes smoothened (smo), fused (fu) and cubitus interruptus (ci)
eliminate wg
transcription at parasegmental borders (Forbes et al., (1993) Development
Suppl. 115-
124; Ingham, (1993) Nature 366:560-562; Preat et al., (1993) Genetics 135:1047-
1062;
and van den Heuvel et al. (1996) Nature 382:547-551); by contrast, mutation of
a fourth
gene, patched (ptc), leads to the derepression of wg (Ingham et al., (1991)
Nature
353:184-187; and Martinez Arias et al., (1988) Development 103:157-170). By
making
double mutant combinations between ptc and the other genes, it was established
that
smo,fu and ci all act downstream of ptc to activate wg transcription (Forbes
et al., (1993)
supra; Hooper (1994) Nature 372:461-464) whilst, on the other hand,
transcription of wg
becomes independent of hh in the absence of ptc (Ingham and Hidalgo (1993)
Development 117:283-291). These findings suggest a simple pathway whereby hh
acts
to antagonize the activity of ptc which in turn antagonizes the activity of
smo, fu and ci.
The universality of this pathway subsequently has been established both in
Drosophila,
where ptc, smo, fu and ci mediate the activity of Hh in all processes studied
to date (Ma
et al., (1993) Cell 75:927-938); Chen et al. (1996) Cell 87:553-563; Forbes et
al., (1996)
Development 122:3283-3294; Sanchez-Herrero et al. (1996) Mech. Dev. 55:159-
170;
Strutt et al. (1997) Development 124:3233-3240), and in vertebrates, where
homologues
of ptc, smo and ci have been identified and implicated in processes mediated
by one or
other of the Hh family proteins (Concordet et al., (1996) Development 122:2835-
2846;
Goodrich et al., supra; Marigo et al., (1996) Dev. Biol. 180:273-283; Stone et
al. (1996)
Nature 384:129-134; Hynes et al. (1997) Neuron 19:15-26; and Quirk et al.
(1997) Cold
Spring Harbor Symp. Quant. Biol. 62:217-226).
Patched was originally identified in Drosophila as a segment polarity gene,
one
of a group of developmental genes that affect cell differentiation within the
individual
segments that occur in a homologous series along the anterior-posterior axis
of the
embryo. See Hooper, J.E. et al. (1989) Cell 59:751; and Nakano, Y. et al.
(1989) Nature
341:508. Patterns of expression of the vertebrate homologue of patched suggest
its
involvement in the development of neural tube, skeleton, limbs, craniofacial
structure,
and skin.
Another protein involved in hedgehog signaling emerged with the discovery that
smoothened also encodes a transmembrane protein that is a member of the 7

CA 02386190 2002-04-11
WO 01/27135 PCT/US00/28479
-8-
transmembrane receptor (7TM) family (Alcedo et al. (1996) Cell 86:221-232; van
den
Heuvel et al. supra). Human homologs of smo have been identified. See, for
example,
Stone et al. (1996) Nature 384:129-134, and GenBank accession U84401. In vitro
binding assays have failed to detect any physical interaction between
vertebrate Smo and
Hh proteins (Stone et al., supra) whereas, under the same conditions,
vertebrate Ptc binds
the Sonic hedgehog (Shh) protein with relatively high affinity (Marigo et al.
(1996)
Nature 384:176-179; Stone et al., supra).Recently, it has been reported that
activating
smoothened mutations occur in sporadic basal cell carcinoma, Xie et al. (1998)
Nature
391: 90-2, and primitive neuroectodermal tumors of the central nervous system,
Reifenberger et al. (1998) Cancer Res 58: 1798-803.
The findings in the art suggest that Hh acts by binding to Ptc, thereby
releasing
an inhibitory effect of Ptc on Smo. Since Ptc and Smo are both transmembrane
proteins,
a proposed scenario is that they physically associate to form a receptor
complex, though
indirect mechanisms of action are also plausible. The derepression of Smo from
Ptc
inhibition most likely involves a conformational change in Smo. It is,
however,
important to remember that Ptc is not essential for Smo's activity, since Smo
becomes
constitutively activated in the complete absence of Ptc protein (Alcedo et
al., supra;
Quirk et al., sgp ra).
It follows from the model that at least some loss-of-function mutations in ptc
should act by disrupting binding to Smo. The discovery that mutations in the
human ptc
homolog are widespread in basal cell carcinomas (BCCs) (Hahn et al. (1996)
Cell
85:841-851; Johnson et al. (1996) Science 272:1668-1671) has provided a major
stimulation for the analysis of Ptc/Smo function as well as an abundant source
of loss-of-
function mutations. Many tumour-derived alleles of human ptc have now been
sequenced, with the majority of the mutations characterized being due to
premature
termination of the coding region (Chidambaram et al. (1996) Cancer Res.
56:4599-4601;
Wicking et al., (1997) Am. J. Hum. Genet. 60:21-26).
Disruption of Smo-Ptc binding could also be caused by mutations in smo; in
contrast to ptc mutations, these should be dominantly acting (since they would
lead to
constitutive activity of the mutant protein). Recent studies of human BCCs
have

CA 02386190 2002-04-11
WO 01/27135 PCT/US00/28479
-9-
identified activating mutation(s) in Smo and appear to be responsible for the
transformation of basal keratinocytes (Xie et al. (1998) Nature 391:90-92).
While not wishing to be bound by any particular theory, the emerging
mechanism by which the smo-ptc pathway mediates signal transduction is as
follows. In
the absence of Hh induction, the activity of Smo is inhibited by Ptc probably
through
their physical association. Full-length Ci forms a complex with Fu, Cos-2 and
suppressor-of-fused [Su(fu)], via which it associates with microtubules. This
association
leads to targeting of Ci to the proteasome where it is cleaved to release the
transcriptional repressing form Ci75. The phosphorylation of Ci155 promotes
its
cleavage, either by promoting association with the Cos-2-Fu or by promoting
ubiquitination (or both). When Hh binds to Ptc, the inhibitory effect on Smo
is
suppressed. The resulting activation of Smo leads to the dissociation of the
Fu-Cos-2-Ci
complex from microtubules. Cleavage of Ci155 is blocked; this or a related
form of Ci
then presumably enters the nucleus to activate transcription of ptc, gli and
other target
genes in association with CREB binding protein (CBP).
Summary of the Invention
One aspect of the present invention makes available methods and reagents for
inhibiting smoothened-dependent pathway activation. In certain embodiments,
the
subject methods can be used to counteract the phenotypic effects of unwanted
activation
of a hedgehog pathway, such as resulting from hedgehog gain-of-function, ptc
loss-of-
function or smoothened gain-of-function mutations. For instance, the subject
method can
involve contacting a cell (in vitro or in vivo) with a smoothened antagonist
(defined
infr a), such as a steroidal alkaloid or other small molecule in an amount
sufficient to
antagonize a smoothened-dependent pathway activation.
Another aspect of the present invention makes available methods and reagents
for activating smoothened-dependent pathway activation, e.g, to mimic all or
certain of
the effects that treatment with a hedgehog protein might cause. The subject
method can
involve contacting a cell (in vitro or in vivo) with a smoothened agonist
(defined infra) in
an amount sufficient to activate a smoothened-dependent pathway.

CA 02386190 2006-08-30
-10-
The subject methods and compounds may be used to regulate proliferation and/or
differentiation of cells in vitro and/or in vivo, e.g., in the formation of
tissue from stem
cells, or to prevent the growth of hyperproliferative cells to illustrate but
a few uses.
The subject compounds may be formulated as a pharmaceutical preparation
comprising a pharmaceutically acceptable excipient. Smoothened antagonists of
the
invention and/or preparations comprising them may be administered to a patient
to treat
conditions involving unwanted cell proliferation, e.g., cancer andJor tumors
(such as
medulloblastoma, basal cell carcinoma, etc.), non-malignant hyperproliferative
disorders, etc. Smoothened agonists can also be used to regulate the growth
and
differentiation of normal tissues. In certain embodiments, such compounds or
preparations are administered systemically and/or locally, e.g., topically.
Brief Description of the Drawings
Figure 1 presents structures of the synthetic compounds AY 9944 and
triparanol,
of the plant steriodal alkaloids jervine, cyclopamine, and tomatidine, and of
cholesterol.
Figure 2 relates to a sensitive assay for Shh signaling in NIH-3T3 cells. (A)
Purification of cholesterol- and palmitate-modified mouse Sonic hedgehog
signaling
domain ShhNp. Detergent-insoluble proteolipid complexes were isolated from 293
cells
expressing full-length Shh (M. K. Cooper, J. A. Porter, K. E. Young, P. A.
Beachy,
Science 280, 1603 (1998)), and ShhNp was purified to apparent homogeneity by
immunoaffinity chromatography. Although recombinant ShhN lacking cholesterol
and
palmitate modifications is fully active in neural plate explant culture
assays, this form of
ShhN was poorly active in the NIH-3T3 cells. We therefore used the detergent
insolubility of cholesterol-modified ShhN and affinity chromatography to
purify the
processed ShhN protein (ShhNp) from a human 293 cell line engineered to
express the
full length mouse Shh construct. Detergent-insoluble complexes (DIGs) were
isolated as
described by Brown and Rose (1992), with the following modifications. Cells
from a
150 mm dish were lysed and ,collected in 2 mL of lysis solution (10 mM NaHPOõ
pH
6.5, 150 mM NaC1, 0.5 mM PMSF, 1% Triton X-100, 2 g/ml Pepstatin A, 10 g/ml
leupeptin, 5 g/ml aprotinin, 2 g/ml E64) at 4 C. Eight sucrose density steps
(35.625-
* Trade-mark

CA 02386190 2006-08-30
-11-
5%, 4.375%/step; made in above solution, without detergent) were layered onto
the 40%
sucrose lysate and centrifugation proceeded for 2-12 hr. Low-density,
flocculent material
was collected (from original position of 18.125% step), diluted 5-fold in 10
mM
NaHPOõ pH 6.5, 150 mM NaCI and harvested by centrifugation at 20,000 x g for
15
min (all at 4 C). Complexes were solubilized at ambient temperature in 1% n-
octyl-a-D-
glucopyranoside, 50 mM HEPES, pH 7.5, 150 mM NaCI and the single ShhNp
immunoaffinity purification step proceeded essentially as described in
Pepinsky et al.
(1998), except the anti-Shh-N monoclonal antibody 5E1 was coupled to Affi-
Gel*10
(Bio-Rad) at 6.6 mg/mI gel. The mass of the purifted species as determined by
mass
spectrometry corresponds to that of the ShhN polypeptide bearing covalent
palmitoyl
and cholesteryl adducts: murine ShhN polypeptide, 19,574.05 Da; esterified
cholesterol,
368.65; palmitoyl mass (in ester or amide linkage), 238.42; sum, 20,181.1. The
inset
shows samples from lysate, detergent-insoluble glycolipid complexes (DIGs; 8
lysate
sample equivalents), and purified ShhNp (0.75 }ig) as separated in SDS-PAGE
(14 !o)
and stained with Coomassie blue. Mass standards migrated as indicated. (B) NIH-
3T3
cells respond to ShhNp. NIH-3T3 cells cotransfected with Gli-luc reporter and
TK
promoter-driven Renilla luciferase control were treated with the indicated
concentrations
of ShhNp for 2 days. Confluent cultures of NIH-3T3 cells were plated at 1:6
dilution to
24 or 96-well plates. On the following day, the cells were transfected with
renilla
luciferase (pRL-TK or pRL-SV40; Clontech) or ji-galactosidase transfection
control
(10% w/w DNA), Gli-Luc reporter (40%) and the constructs indicated (50 l0)
using
Fugene 6 (Roche) transfection reagent (250 ng (24 well plate) or 100 ng (96
well plate)
DNA/we11, 3:1 ratio (v/w) of reagent to DNA). After the cells had reached
saturation
density (1-2 d), they were changed to low serum medium (0.5% bovine calf
serum), and
treated with the reagents indicated for 1-2 d. Firefly and Renilla luciferase
and p-
galactosidase activities were assayed from the cell lysates Iiy luminometry
using dual
~
luciferase (Promega) and Galacto-Light (Tropix) kits, respectively. Luciferase
activities
are normalized relative to control; a representative experiment is shown. Note
that in this
and all subsequent reporter assays, TK-Renilla luciferase activity is used as
a control for
normalization. (C) Shh pathway activation is sensitive to cyclopamine in NIH-
3T3 cells.
NIH-3T3 cells transfected as above (in triplicate) were treated with ShhNp (4
nM) and/or
*Trade-mark

CA 02386190 2002-04-11
WO 01/27135 PCT/USOO/28479
-12-
cyclopamine (5 M) for 2 d as indicated. Normalized luciferase activities are
given as
fold induction relative to control. Error bars indicate one standard
deviation. (D) Low
cell density inhibits Shh pathway activity downstream of Smo. Cultures of Shh-
LIGHT
(open boxes) or SmoA 1-LIGHT (filled diamonds) cells were plated to 96-well
plates in a
series of twofold dilutions. The NIH-3T3 cell clone Shh-LIGHT and Shh-LIGHT2
stably incorporating the Gli-luc reporter and TK-renilla vectors were
established by
cotransfection with a vector encoding G418 resistance (pSV-Neo), followed by
selection
with G418 and cell cloning. Subsequently, a clonal subline of Shh-LIGHT
expressing
activated Smo (SmoAl-LIGHT) was established using an expression vector that
allows
hygromycin selection (pcDNA3.1+ hygro; Invitrogen). The expression of SmoAl in
the
cell line was verified by immunoblotting. The Shh-LIGHT cells were treated
with 4 nM
ShhNp, and Gli-luciferase reporter activity was assayed after 24 h. Fold
induction of the
reporter (% of maximum is relative to equally dense Shh-LIGHT control culture)
and
cell densities (% of maximum Renilla luciferase activity) were measured at the
end of
the experiment. Error bars indicate one standard deviation (quadruplicate
wells).
Figure 3 demonstrates how cyclopamine acts by inhibiting the activity of Smo.
(A) Ptcl-/- cells are sensitive to cyclopamine. Fibroblasts from Ptcl-/-
embryos were
treated with cyclopamine or forskolin as indicated. After 3 d, cells were
lysed and ~3-
galactosidase activity relative to protein concentration was measured. Since
(3-
galactosidase is expressed from the Ptcl locus, its expression reflects the
activity of the
Shh pathway. A representative experiment is shown. (B) Activated mutants of
Smo are
resistant to cyclopamine. Cultures of NIH-3T3 cells were transfected (in
triplicate) with
Gli-luciferase reporter, TK-Renilla luciferase control vector and Smo or SmoAl
expression vectors. Smo DNA was used at 50% w/w, and SmoAl at 50%, 5%, and
0.5%
w/w. Subsequently, the cultures were treated with 5 M cyclopamine for 2 d.
Error bars
indicate one standard deviation. The leftmost four bars, shown for comparison,
are as in
Fig. 2C. (C) High level expression of Ptcl restores cyclopamine resistant
response of
SmoAl to ShhNp, NIH-3T3 cells were transfected with Gli-luc reporter, TK-
Renilla,
Ptc1CTD and SmoAl expression vectors (Ptc to Smo DNA ratio = 9). Subsequently,
the
cultures were treated with ShhNp (2 nM), cyclopamine (5 M) and/or forskolin
(100
M) as indicated for 2 d. Note that SmoAl activation of pathway is dramatically
reduced by high levels of Ptcl activity (compare to panel B), and that 2 nM
ShhNp

CA 02386190 2006-08-30
-13-
restores pathway activity even. in the presence of 5 M cyclopamine. A
representative
experiment is shown. (D) Tumor-derived mutant Smo proteins are intrinsically
more
active than wild type Smo. NIH-3T3 cells were cotransfected with Gli-luc
reporter, (3-
galactosidase transfection control, and a control vector or an expression
vector encoding
the indicated Smo-Renilla luciferase fusion protein. In the representative
experiment
shown, Shh pathway activity and Smo protein levels were measured as firefly
and
Renilla luciferase activities relative to 0-galactosidase, respectively.
Epitope-tagged Smo
and activated Smo proteins also displayed similar levels of expression in
these cells (not
shown).
Figure 4 depicts a cyclopamine derivative of increased potency. (A) 3-Keto, N-
aminoethyl aminocaproyl dihydrocinnamoyl cyclopamine (KAAD cyclopamine) was
synthesized from cyclopamine. Structure of KAAD cyclopamine was verified by
NMR
and mass spectrometry analyses. KAAD cyclopamine can block pathway activation
by
tumor-derived Smo. Shh-LIGHT2 (diamonds) and SmoAl-LIGHT (circles) cells were
treated with 4 nM ShhNp (Shh-LIGHT2) and increasing concentrations of KAAD
cyclopamine (both lines) for 2 d. Relative reporter activity is normalized to
maximum.
Note the increased inhibitory potency of KAAD cyclopamine as compared to
cyclopamine in Fig. 3 A-C. (B) KAAD cyclopamine can block pathway activation
in
Ptcl-/- cells. p2PTC_/- cells (these cells are a cloned line derived from Ptc-
/- mouse
embryonic fibroblasts) were treated with increasing concentrations of
cyclopamine (open
boxes) or KAAD cyclopamine (filled boxes) for 2d. The suppression of pathway
activity
induced by SmoAl-Renilla by high concentrations of cyclopamine derivatives did
not
involve a decrease in the level of expression of the Smo construct (not
shown). Cells
were seeded into duplicate 96-well plates, allowed to grow to saturation
density, and
incubated with cyclopamine and KAAD cyclopamine for 2 d. (3-galactosidase
activity
was determined using Galacto-Light kit (no inactivation of endogenous P-gal
activity,
Tropix). P-galactosidase activities were normalized to cell mass as determined
from a
treated duplicate plate using the Cell Titer 96AQ* assay (Promega). Maximum
normalized P-galactosidase activities (1103 for KAAD-cyclopamine and 916 for
cyclopamine) were set to 1 and minimum activities (191 and 144, respectively)
were set
to 0. Significant toxicity (microscopically visible cell death, or decrease in
Cell Titer
reading) was not observed, even at the highest doses of compounds used. 0-
galactosidase
*Trade-mark

CA 02386190 2002-04-11
WO 01/27135 PCT/US00/28479
-14-
activity is normalized to the maximum. Error bars in A and B indicate one
standard
deviation.
Figure 5 presents inhibitors of the Hedgehog pathway according to the present
invention.
Figure 6 depicts response of fibrosarcoma tumors to treatment with a subject
compound.
Figure 7 compares tumor tissue following treatment with tomatidine with tissue
treated with a subject compound.
Detailed Description of the Invention
I. Overview
The present invention relates to the discovery that signal transduction
pathways
regulated by patched (ptc) and/or smoothened can be inhibited, at least in
part, by
steroidal alkaloids and analogs thereof. As set out in more detail below, we
have
observed that derivatives of cyclopamine can inhibit smoothened-dependent
activity of
the hedgehog pathway. While not wishing to be bound by any particular theory,
our data
indicates that cyclopamine acts at the level of smoothened, directly or
indirectly shifting
the steady-state ratio of active and inactive forms of smoothened towards the
inactive
form (e.g., relative to the absence of the steroidal alkaloid).
It is, therefore, specifically contemplated that other small molecules,
steroidal
and non-steroidal in structure, may similarly interfere with aspects of
smoothened-
mediated signal transduction. For instance, such compounds may be useful for
inhibiting
proliferation and/or inducing differentiation of normal tissues (e.g., tissues
which
express smo or are otherwise hedgehog-responsive). The subject smoothened
antagonists
may also be used to inhibit proliferation (or other biological consequences)
in cells or
tissues characterized as having a patched loss-of-function phenotype, a
smoothened
gain-of-function phenotype or a hedgehog gain-of-function phenotype.
It is also specifically contemplated that, in light of cyclopamine and other
small
molecules being able to inhibit smoothened-mediated signal transduction, that
activators
of smoothened-mediated signal transduction can be identified, e.g., compounds
which

CA 02386190 2006-08-30
-15-
directly or indirectly shift the steady-state ratio of active and inactive
forms of
smoothened towards the active form. Such compounds may be useful for, to
illustrate,
inducing proliferation and/or preventing differentiation of normal tissues
(e.g., tissues
which express smo or are otherwise hedgehog-responsive).
In preferred embodiments, the subject inhibitors and activators are organic
molecules having a molecular weight less than 2500 amu, more preferably less
than
1500 amu, and even more preferably less than 750 amu, and are capable of
inhibiting at
least some of the activity of a smoothened signal transduction pathway.
Thus, the methods of the present invention include the use of agents, such as
small molecules, which antagonize or activate (as appropriate) smoothened-
dependent
activity of the hedgehog pathway, resulting in the regulation of repair and/or
functional
performance of a wide range of cells, tissues, and organs. For instance, the
subject
methods have therapeutic and cosmetic applications ranging from regulation of
neural
tissues, bone and cartilage formation and repair, regulation of
spermatogenesis,
regulation of smooth muscle, regulation of lung, liver, pancreas, and other
organs arising
from the primitive gut, regulation of hematopoietic function, regulation of
skin and hair
growth, etc. Moreover, the subject methods can be performed on cells which are
provided in culture (in vitro), or on cells in a whole animal (in vivo). See,
for example,
PCT publications WO 95/18856 and WO 96/17924.
In a certain preferred embodiment, the subject smoothened antagonists can be
to
treat epithelial cells having a phenotype of ptc loss-of-function, hedgehog
gain-of-
function, or smoothened gain-of-function employing an agent which antagonizes
hedgehog function. For instance, the subject method can be used in treating or
preventing basal cell carcinoma or other hedgehog pathway-related disorders.
In another preferred embodiment, the subject smoothened antagonists and
activators can, as appropriate, be used to modulate proliferation or
differentiation of
pancreatic cells (e.g., ranging from pancreatic progenitor cells and mature
endocrine or
exocrine cells), or to regulate the growth or development of pancreatic
tissue, e.g., in
vivo or in vitro.

CA 02386190 2002-04-11
WO 01/27135 PCT/US00/28479
-16-
In yet another preferred embodiment, the subject method can be used as part of
a
treatment regimen for malignant medulloblastoma and other primary CNS
malignant
neuroectodermal tumors.
In another aspect, the present invention provides pharmaceutical preparations
comprising, as an active ingredient, a smoothened antagonist or activator such
as
described herein, formulated in an amount sufficient to regulate, in vivo, a
smoothened-
dependent pathway, e.g., proliferation, differentiation or other biological
consequences
of normal or abnormal function of, for example, ptc, hedgehog or smoothened.
The subject treatments using the subject compounds can be effective for both
human and animal subjects. Animal subjects to which the invention is
applicable extend
to both domestic animals and livestock, raised either as pets or for
commercial purposes.
Examples are dogs, cats, cattle, horses, sheep, hogs, and goats.
H. Definitions
For convience, certain terms employed in the specification, examples, and
appended claims are collected here.
The phrase "aberrant modification or mutation" of a gene refers to such
genetic
lesions as, for example, deletions, substitution or addition of nucleotides to
a gene, as
well as gross chromosomal rearrangements of the gene and/or abnormal
methylation of
the gene. Likewise, mis-expression of a gene refers to aberrant levels of
transcription of
the gene relative to those levels in a normal cell under similar conditions,
as well as non-
wild-type splicing of mRNA transcribed from the gene.
"Basal cell carcinomas" exist in a variety of clinical and histological forms
such
as nodular-ulcerative, superficial, pigmented, morphealike, fibroepithelioma
and nevoid
syndrome. Basal cell carcinomas are the most common cutaneous neoplasms found
in
humans. The majority of new cases of nonmelanoma skin cancers fall into this
category.
"Burn wounds" refer to cases where large surface areas of skin have been
removed or lost from an individual due to heat and/or chemical agents.

CA 02386190 2002-04-11
WO 01/27135 PCT/US00/28479
-17-
The term "cAMP regulator" refers to an agent which alters the level or
activity of
cAMP in a cell, including agents which act upon adenylate cyclase, cAMP
phosphodiesterase, or other molecules which, in turn, regulate cAMP levels or
activity.
Additionally, cAMP regulators, as the term is used herein, refer to downstream
effectors
of cAMP activity, such as protein kinase A. "cAMP agonists" refers. to that
subset of
cAMP regulators which increases the level or activity of cAMP in a cell, while
"cAMP
antagonists" refers to the subset which decreases the level or activity of
cAMP in a cell.
The term "carcinoma" refers to a malignant new growth made up of epithelial
cells tending to infiltrate surrounding tissues and to give rise to
metastases. Exemplary
carcinomas include: "basal cell carcinoma", which is an epithelial tumor of
the skin that,
while seldom metastasizing, has potentialities for local invasion and
destruction;
"squamous cell carcinoma", which refers to carcinomas arising from squamous
epithelium and having cuboid cells; "carcinosarcoma", which include malignant
tumors
composed of carcinomatous and sarcomatous tissues; "adenocystic carcinoma",
carcinoma marked by cylinders or bands of hyaline or mucinous stroma separated
or
surrounded by nests or cords of small epithelial cells, occurring in the
mammary and
salivary glands, and mucous glands of the respiratory tract; "epidermoid
carcinoma",
which refers to cancerous cells which tend to differentiate in the same way as
those of
the epidermis; i.e., they tend to form prickle cells and undergo comification;
"nasopharyngeal carcinoma", which refers to a malignant tumor arising in the
epithelial
lining of the space behind the nose; and "renal cell carcinoma", which
pertains to
carcinoma of the renal parenchyma composed of tubular cells in varying
arrangements.
Other carcinomatous epithelial growths are "papillomas", which refers to
benign tumors
derived from epithelium and having a papillomavirus as a causative agent; and
"epidermoidomas", which refers to a cerebral or meningeal tumor formed by
inclusion of
ectodermal elements at the time of closure of the neural groove.
The "corium" or "dermis" refers to the layer of the skin deep to the
epidermis,
consisting of a dense bed of vascular connective tissue, and containing the
nerves and
terminal organs of sensation. The hair roots, and sebaceous and sweat glands
are
structures of the epidermis which are deeply embedded in the dermis.

CA 02386190 2002-04-11
WO 01/27135 PCT/US00/28479
-18-
"Dental tissue" refers to tissue in the mouth which is similar to epithelial
tissue,
for example gum tissue. The method of the present invention is useful for
treating
periodontal disease.
"Dermal skin ulcers" refer to lesions on the skin caused by superficial loss
of
tissue, usually with inflammation. Dermal skin ulcers which can be treated by
the
method of the present invention include decubitus ulcers, diabetic ulcers,
venous stasis
ulcers and arterial ulcers. Decubitus wounds refer to chronic ulcers that
result from
pressure applied to areas of the skin for extended periods of time. Wounds of
this type
are often called bedsores or pressure sores. Venous stasis ulcers result from
the
stagnation of blood or other fluids from defective veins. Arterial ulcers
refer to necrotic
skin in the area around arteries having poor blood flow.
The term "ED50" means the dose of a drug which produces 50% of its maximum
response or effect.
An "effective amount" of a subject compound, with respect to the subject
method
of treatment, refers to an amount of the antagonist in a preparation which,
when applied
as part of a desired dosage regimen brings about, e.g., a change in the rate
of cell
proliferation and/or the state of differentiation of a cell and/or rate of
survival of a cell
according to clinically acceptable standards for the disorder to be treated or
the cosmetic
purpose.
The terms "epithelia", "epithelial" and "epithelium" refer to the cellular
covering
of internal and external body surfaces (cutaneous, mucous and serous),
including the
glands and other structures derived therefrom, e.g., corneal, esophogeal,
epidermal, and
hair follicle epithelial cells. Other exemplary epithlelial tissue includes:
olfactory
epithelium, which is the pseudostratified epithelium lining the olfactory
region of the
nasal cavity, and containing the receptors for the sense of smell; glandular
epithelium,
which refers to epithelium composed of secreting cells; squamous epithelium,
which
refers to epithelium composed of flattened plate-like cells. The term
epithelium can also
refer to transitional epithelium, like that which is characteristically found
lining hollow
organs that are subject to great mechanical change due to contraction and
distention, e.g.,
tissue which represents a transition between stratified squamous and columnar
epithelium.

CA 02386190 2002-04-11
WO 01/27135 PCT/USOO/28479
-19-
The term "epithelialization" refers to healing by the growth of epithelial
tissue
over a denuded surface.
The term "epidermal gland" refers to an aggregation of cells associated with
the
epidermis and specialized to secrete or excrete materials not related to their
ordinary
metabolic needs. For example, "sebaceous glands" are holocrine glands in the
corium
that secrete an oily substance and sebum. The term "sweat glands" refers to
glands that
secrete sweat, situated in the corium or subcutaneous tissue, opening by a
duct on the
body surface.
The term "epidermis" refers to the outermost and nonvascular layer of the
skin,
derived from the embryonic ectoderm, varying in thickness from 0.07-1.4 mm. On
the
palmar and plantar surfaces it comprises, from within outward, five layers:
basal layer
composed of columnar cells arranged perpendicularly; prickle-cell or spinous
layer
composed of flattened polyhedral cells with short processes or spines;
granular layer
composed of flattened granular cells; clear layer composed of several layers
of clear,
transparent cells in which the nuclei are indistinct or absent; and horny
layer composed
of flattened, cornified non-nucleated cells. In the epidermis of the general
body surface,
the clear layer is usually absent.
"Excisional wounds" include tears, abrasions, cuts, punctures or lacerations
in
the epithelial layer of the skin and may extend into the dermal layer and even
into
subcutaneous fat and beyond. Excisional wounds can result from surgical
procedures or
from accidental penetration of the skin.
The "growth state" of a cell refers to the rate of proliferation of the cell
and/or
the state of differentiation of the cell. An "altered growth state" is a
growth state
characterized by an abnormal rate of proliferation, e.g., a cell exhibiting an
increased or
decreased rate of proliferation relative to a normal cell.
The term "hair" refers to a threadlike structure, especially the specialized
epidermal structure composed of keratin and developing from a papilla sunk in
the
corium, produced only by mammals and characteristic of that group of animals.
Also,
"hair" may refer to the aggregate of such hairs. A "hair follicle" refers to
one of the
tubular-invaginations of the epidermis enclosing the hairs, and from which the
hairs

CA 02386190 2002-04-11
WO 01/27135 PCT/USOO/28479
-20-
grow. "Hair follicle epithelial cells" refers to epithelial cells which
surround the dermal
papilla in the hair follicle, e.g., stem cells, outer root sheath cells,
matrix cells, and inner
root sheath cells. Such cells may be normal non-malignant cells, or
transformed/immortalized cells.
The term "smoothened antagonist" refers to an agent which represses or induces
transcription of target genes, e.g., glil and ptc genes, which in normal cells
are induced
or repressed by contact of the cell with hedgehog. In addition to altering a
smoothened-
dependent pathway, preferred smoothened antagonists can be used to overcome a
ptc
loss-of-function and/or a smoothened gain-of-function. The term "smoothened
antagonist" as used herein also refers to any agent that may act by regulating
a
downstream effector of the smoothened pathway such as fused, suppressor of
fused,
cubitus interruptus, costal-2, etc., thereby inhibiting smoothened-dependent
pathway
activation.
The terms "loss-of-function" and "gain-of-function", as appropriate, refer to
an
aberrant modification or mutation of, e.g., a ptc gene, hedgehog gene, or
smoothened
gene, or a decrease or increase in the level of expression of such a gene,
which results in
a phenotype, e.g., which resembles contacting a cell with a hedgehog protein,
such as
aberrant activation of a hedgehog pathway or resemble loss of smo function.
The
mutation may include a loss of the ability of the ptc or smo gene product(s)
to regulate
the level of activity of Ci proteins, e.g., Glfl, Gli2, and Gli3.
As used herein, "immortalized cells" refers to cells which have been altered
via
chemical and/or recombinant means such that the cells have the ability to grow
through
an indefinite number of divisions in culture.
"Internal epithelial tissue" refers to tissue inside the body which has
characteristics similar to the epidermal layer in the skin. Examples include
the lining of
the intestine. The method of the present invention is useful for promoting the
healing of
certain internal wounds, for example wounds resulting from surgery.
The term "keratosis" refers to proliferative skin disorder characterized by
hyperplasia of the horny layer of the epidermis. Exemplary keratotic disorders
include

CA 02386190 2002-04-11
WO 01/27135 PCT/US00/28479
-21-
keratosis follicularis, keratosis palmaris et plantaris, keratosis pharyngea,
keratosis
pilaris, and actinic keratosis.
The term "LD50" means the dose of a drug which is lethal in 50% of test
subjects.
The term "nail" refers to the horny cutaneous plate on the dorsal surface of
the
distal end of a finger or toe.
A "patient" or "subject" to be treated by the subject method can mean either a
human or non-human animal.
The term "prodrug" is intended to encompass compounds which, under
physiological conditions, are converted into the therapeutically active agents
of the
present invention. A common method for making a prodrug is to include selected
moieties which are hydrolyzed under physiological conditions to reveal the
desired
molecule. In other embodiments, the prodrug is converted by an enzymatic
activity of
the host animal.
As used herein, "proliferating" and "proliferation" refer to cells undergoing
mitosis.
Throughout this application, the term "proliferative skin disorder" refers to
any
disease/disorder of the skin marked by unwanted or aberrant proliferation of
cutaneous
tissue. These conditions are typically characterized by epidermal cell
proliferation or
incomplete cell differentiation, and include, for example, X-linked
ichthyosis, psoriasis,
atopic dermatitis, allergic contact dermatitis, epidermolytic hyperkeratosis,
and
seborrheic dermatitis. For example, epidermodysplasia is a form of faulty
development
of the epidermis. Another example is "epidermolysis", which refers to a
loosened state of
the epidermis with formation of blebs and bullae either spontaneously or at
the site of
trauma.
As used herein, the term "psoriasis" refers to a hyperproliferative skin
disorder
which alters the skin's regulatory mechanisms. In particular, lesions are
formed which
involve primary and secondary alterations in epidermal proliferation,
inflammatory
responses of the skin, and an expression of regulatory molecules such as
lymphokines
and inflammatory factors. Psoriatic skin is morphologically characterized by
an

CA 02386190 2002-04-11
WO 01/27135 PCT/US00/28479
-22-
increased turnover of epidermal cells, thickened epidermis, abnormal
keratinization,
inflammatory cell infiltrates into the dermis layer and polymorphonuclear
leukocyte
infiltration into the epidermis layer resulting in an increase in the basal
cell cycle.
Additionally, hyperkeratotic and parakeratotic cells are present.
The term "skin" refers to the outer protective covering of the body,
consisting of
the corium and the epidermis, and is understood to include sweat and sebaceous
glands,
as well as hair follicle structures. Throughout the present application, the
adjective
"cutaneous" may be used, and should be understood to refer generally to
attributes of the
skin, as appropriate to the context in which they are used.
The term "therapeutic index" refers to the therapeutic index of a drug defined
as
LD50/ED50=
As used herein, "transformed cells" refers to cells which have spontaneously
converted to a state of unrestrained growth, i.e., they have acquired the
ability to grow
through an indefinite number of divisions in culture. Transformed cells may be
characterized by such terms as neoplastic, anaplastic and/or hyperplastic,
with respect to
their loss of growth control.
The term "acylamino" is art-recognized and refers to a moiety that can be
represented by the general formula:
O
_ II R
'ii
i
R9
wherein R9 is as defined above, and R' 11 represents a hydrogen, an alkyl, an
alkenyl or
-(CH2)m-R8, wherein R8 represents an aryl, a cycloalkyl, a cycloalkenyl, a
heterocycle
or a polycycle; and m is zero or an integer in the range of 1 to 8.
Herein, the term "aliphatic group" refers to a straight-chain, branched-chain,
or
cyclic aliphatic hydrocarbon group and includes saturated and unsaturated
aliphatic
groups, such as an alkyl group, an alkenyl group, and an alkynyl group.

CA 02386190 2002-04-11
WO 01/27135 PCT/US00/28479
- 23 -
The terms "alkenyl" and "alkynyl" refer to unsaturated aliphatic groups
analogous in length and possible substitution to the alkyls described above,
but that
contain at least one double or triple bond respectively.
The terms "alkoxyl" or "alkoxy" as used herein refers to an alkyl group, as
defined above, having an oxygen radical attached thereto. Representative
alkoxyl groups
include methoxy, ethoxy, propyloxy, tert-butoxy and the like. An "ether" is
two
hydrocarbons covalently linked by an oxygen. Accordingly, the substituent of
an alkyl
that renders that alkyl an ether is or resembles an alkoxyl, such as can be
represented by
one of -0-alkyl, -0-alkenyl, -0-alkynyl, -0-(CH2)m-R8, where m and R8 are
described
above.
The term "alkyl" refers to the radical of saturated aliphatic groups,
including
straight-chain alkyl groups, branched-chain alkyl groups, cycloalkyl
(alicyclic) groups,
alkyl-substituted cycloalkyl groups, and cycloalkyl-substituted alkyl groups.
In preferred
embodiments, a straight chain or branched chain alkyl has 30 or fewer carbon
atoms in
its backbone (e.g., Cl-C30 for straight chains, C3-C30 for branched chains),
and more
preferably 20 or fewer. Likewise, preferred cycloalkyls have from 3-10 carbon
atoms in
their ring structure, and more preferably have 5, 6 or 7 carbons in the ring
structure.
Moreover, the term "alkyl" (or "lower alkyl") as used throughout the
specification, examples, and claims is intended to include both "unsubstituted
alkyls"
and "substituted alkyls", the latter of which refers to alkyl moieties having
substituents
replacing a hydrogen on one or more carbons of the hydrocarbon backbone. Such
substituents can include, for example, a halogen, a hydroxyl, a carbonyl (such
as a
carboxyl, an alkoxycarbonyl, a formyl, or an acyl), a thiocarbonyl (such as a
thioester, a
thioacetate, or a thioformate), an alkoxyl, a phosphoryl, a phosphate, a
phosphonate, a
phosphinate, an amino, an amido, an amidine, an imine, a cyano, a nitro, an
azido, a
sulfhydryl, an alkylthio, a sulfate, a sulfonate, a sulfamoyl, a sulfonamido,
a sulfonyl, a
heterocyclyl, an aralkyl, or an aromatic or heteroaromatic moiety. It will be
understood
by those skilled in the art that the moieties substituted on the hydrocarbon
chain can
themselves be substituted, if appropriate. For instance, the substituents of a
substituted
alkyl may include substituted and unsubstituted forms of amino, azido, imino,
amido,
phosphoryl (including phosphonate and phosphinate), sulfonyl (including
sulfate,

CA 02386190 2002-04-11
WO 01/27135 PCT/USOO/28479
-24-
sulfonamido, sulfamoyl and sulfonate), and silyl groups, as well as ethers,
alkylthios,
carbonyls (including ketones, aldehydes, carboxylates, and esters), -CF3, -CN
and the
like. Exemplary substituted alkyls are described below. Cycloalkyls can be
further
substituted with alkyls, alkenyls, alkoxys, alkylthios, aminoalkyls, carbonyl-
substituted
alkyls, -CF3, -CN, and the like.
Unless the number of carbons is otherwise specified, "lower alkyl" as used
herein
means an alkyl group, as defined above, but having from one to ten carbons,
more
preferably from one to six carbon atoms in its backbone structure. Likewise,
"lower
alkenyl" and "lower alkynyl" have similar chain lengths. Throughout the
application,
preferred alkyl groups are lower alkyls. In preferred embodiments, a
substituent
designated herein as alkyl is a lower alkyl.
The term "alkylthio" refers to an alkyl group, as defined above, having a
sulfur
radical attached thereto. In preferred embodiments, the "alkylthio" moiety is
represented
by one of -S-alkyl, -S-alkenyl, -S-alkynyl, and -S-(CH2)m-R8, wherein m and R8
are
defined above. Representative alkylthio groups include methylthio, ethylthio,
and the
like.
The terms "amine" and "amino" are art-recognized and refer to both
unsubstituted and substituted amines, e.g., a moiety that can be represented
by the
general formula:
Rtio
~Rio 1+
-N or - i -Rio
R9 R
9
wherein Rg, RI O and R' 10 each independently represent a hydrogen, an alkyl,
an
alkenyl, -(CH2)m-R8, or R9 and R10 taken together with the N atom to which
they are
attached complete a heterocycle having from 4 to 8 atoms in the ring
structure; R8
represents an aryl, a cycloalkyl, a cycloalkenyl, a heterocycle or a
polycycle; and m is
zero or an integer in the range of 1 to 8. In preferred embodiments, only one
of R9 or
RIO can be a carbonyl, e.g., R9, R10 and the nitrogen together do not form an
imide. In
even more preferred embodiments, Rg and RI 0(and optionally R' 10) each

CA 02386190 2002-04-11
WO 01/27135 PCT/US00/28479
-25-
independently represent a hydrogen, an alkyl, an alkenyl, or -(CH2)m-R8. Thus,
the term
"alkylamine" as used herein means an amine group, as defined above, having a
substituted or unsubstituted alkyl attached thereto, i.e., at least one of R9
and R10 is an
alkyl group.
The term "amido" is art-recognized as an amino-substituted carbonyl and
includes a moiety that can be represented by the general formula:
O
N __R9
R io
wherein R9, R10 are as defined above. Preferred embodiments of the amide will
not
include imides which may be unstable.
The term "aralkyl", as used herein, refers to an alkyl group substituted with
an
aryl group (e.g., an aromatic or heteroaromatic group).
The term "aryl" as used herein includes 5-, 6-, and 7-membered single-ring
aromatic groups that may include from heteroatoms (preferably 1 to 4), for
example,
benzene, pyrrole, furan, thiophene, imidazole, oxazole, thiazole, triazole,
pyrazole,
pyridine, pyrazine, pyridazine and pyrimidine, and the like. Those aryl groups
having
heteroatoms in the ring structure may also be referred to as "aryl
heterocycles" or
"heteroaromatics." The aromatic ring can be substituted at one or more ring
positions
with such substituents as described above, for example, halogen, azide, alkyl,
aralkyl,
alkenyl, alkynyl, cycloalkyl, hydroxyl, alkoxyl, amino, nitro, sulfhydryl,
imino, amido,
phosphate, phosphonate, phosphinate, carbonyl, carboxyl, silyl, ether,
alkylthio,
sulfonyl, sulfonamido, ketone, aldehyde, ester, heterocyclyl, aromatic or
heteroaromatic
moieties, -CF3, -CN, or the like. The term "aryl" also includes polycyclic
ring systems
having two or more cyclic rings in which two or more carbons are common to two
adjoining rings (the rings are "fused rings") wherein at least one of the
rings is aromatic,
e.g., the other cyclic rings can be cycloalkyls, cycloalkenyls, cycloalkynyls,
aryls and/or
heterocyclyls.

CA 02386190 2002-04-11
WO 01/27135 PCT/US00/28479
-26-
The term "carbocycle", as used herein, refers to an aromatic or non-aromatic
ring
in which each atom of the ring is carbon.
The term "carbonyl" is art-recognized and includes such moieties as can be
represented by the general formula:
0 0
~X-Rii , or-X~R ,
11
wherein X is a bond or represents an oxygen or a sulfur, and R1 I represents a
hydrogen,
an alkyl, an alkenyl, -(CH2)m-R8 or a pharmaceutically acceptable salt, R' 11
represents
a hydrogen, an alkyl, an alkenyl or -(CH2)m-R8, where m and R8 are as defined
above.
Where X is an oxygen and RlI or R'11 is not hydrogen, the formula represents
an
"ester". Where X is an oxygen, and Ri1 is as defined above, the moiety is
referred to
herein as a carboxyl group, and particularly when Rll is a hydrogen, the
formula
represents a "carboxylic acid". Where X is an oxygen, and R'I 1 is hydrogen,
the formula
represents a "formate". In general, where the oxygen atom of the above formula
is
replaced by sulfur, the formula represents a "thiocarbonyl" group. Where X is
a sulfur
and RI 1 or R' 11 is not hydrogen, the formula represents a "thioester." Where
X is a
sulfur and RI 1 is hydrogen, the formula represents a "thiocarboxylic acid."
Where X is a
sulfur and RI 1' is hydrogen, the formula represents a "thiolformate." On the
other hand,
where X is a bond, and R1 I is not hydrogen, the above formula represents a
"ketone"
group. Where X is a bond, and RII is hydrogen, the above formula represents an
"aldehyde" group.
The term "heteroatom" as used herein means an atom of any element other than
carbon or hydrogen. Preferred heteroatoms are boron, nitrogen, oxygen,
phosphorus,
sulfur and selenium.
The terms "heterocyclyl" or "heterocyclic group" refer to 3- to 10-membered
ring
structures, more preferably 3- to 7-membered rings, whose ring structures
include one to
four heteroatoms. Heterocycles can also be polycycles. Heterocyclyl groups
include, for
example, thiophene, thianthrene, furan, pyran, isobenzofuran, chromene,
xanthene,

CA 02386190 2002-04-11
WO 01/27135 PCT/US00/28479
-27-
phenoxathiin, pyrrole, imidazole, pyrazole, isothiazole, isoxazole, pyridine,
pyrazine,
pyrimidine, pyridazine, indolizine, isoindole, indole, indazole, purine,
quinolizine,
isoquinoline, quinoline, phthalazine, naphthyridine, quinoxaline, quinazoline,
cinnoline,
pteridine, carbazole, carboline, phenanthridine, acridine, pyrimidine,
phenanthroline,
phenazine, phenarsazine, phenothiazine, furazan, phenoxazine, pyrrolidine,
oxolane,
thiolane, oxazole, piperidine, piperazine, morpholine, lactones, lactams such
as
azetidinones and pyrrolidinones, sultams, sultones, and the like. The
heterocyclic ring
can be substituted at one or more positions with such substituents as
described above, as
for example, halogen, alkyl, aralkyl, alkenyl, alkynyl, cycloalkyl, hydroxyl,
amino, nitro,
sulfhydryl, imino, amido, phosphate, phosphonate, phosphinate, carbonyl,
carboxyl,
silyl, ether, alkylthio, sulfonyl, ketone, aldehyde, ester, a heterocyclyl, an
aromatic or
heteroaromatic moiety, -CF3, -CN, or the like.
As used herein, the term "nitro" means -NO2; the term "halogen" designates -F,
-
Cl, -Br or -I; the term "sulfhydryP" means -SH; the term "hydroxyl" means -OH;
and the
term "sulfonyl" means -SO2-.
A"phosphonamidite" can be represented in the general formula:
R48 R48
-Qz i-O ' or QZ i- OR46
N (Ry) Rlo N (Ry) Rlo
wherein Ry and R10 are as defined above, Q2 represents 0, S or N, and R48
represents a
lower alkyl or an aryl, Q2 represents 0, S or N.
A "phosphoramidite" can be represented in the general formula:
0 0
ii u
-Qz i -O- -QZ P- OR46
, or 1
N (R9) R10 N (R9) Rlo
wherein R9 and R10 are as defined above, and Q2 represents 0, S or N.
A "phosphoryl" can in general be represented by the formula:

CA 02386190 2002-04-11
WO 01/27135 PCT/US00/28479
-28-
41
II
-p-
1
OR4 6
wherein Ql represented S or 0, and R46 represents hydrogen, a lower alkyl or
an aryl.
When used to substitute, for example, an alkyl, the phosphoryl group of the
phosphorylalkyl can be represented by the general formula:
41 41
II II
-QZ p-0- -QZ p- OR46
I , or I
OR46 OR4 6
wherein Q1 represented S or 0, and each R46 independently represents hydrogen,
a
lower alkyl or an aryl, Q2 represents 0, S or N. When Q 1 is an S, the
phosphoryl moiety
is a "phosphorothioate".
The terms "polycyclyl" or "polycyclic group" refer to two or more rings (e.g.,
cycloalkyls, cycloalkenyls, cycloalkynyls, aryls and/or heterocyclyls) in
which two or
more carbons are common to two adjoining rings, e.g., the rings are "fused
rings". Rings
that are joined through non-adjacent atoms are termed "bridged" rings. Each of
the rings
of the polycycle can be substituted with such substituents as described above,
as for
example, halogen, alkyl, aralkyl, alkenyl, alkynyl, cycloalkyl, hydroxyl,
amino, nitro,
sulfhydryl, imino, amido, phosphate, phosphonate, phosphinate, carbonyl,
carboxyl,
silyl, ether, alkylthio, sulfonyl, ketone, aldehyde, ester, a heterocyclyl, an
aromatic or
heteroaromatic moiety, -CF3, -CN, or the like.
The phrase "protecting group" as used herein means temporary substituents
which protect a potentially reactive functional group from undesired chemical
transformations. Examples of such protecting groups include esters of
carboxylic acids,
silyl ethers of alcohols, and acetals and ketals of aldehydes and ketones,
respectively.
The field of protecting group chemistry has been reviewed (Greene, T.W.; Wuts,
P.G.M.
Protective Groups in Organic Synthesis, 2 d ed.; Wiley: New York, 1991).
A "selenoalkyl" refers to an alkyl group having a substituted seleno group
attached thereto. Exemplary "selenoethers" which may be substituted on the
alkyl are

CA 02386190 2002-04-11
WO 01/27135 PCT/US00/28479
-29-
selected from one of -Se-alkyl, -Se-alkenyl, -Se-alkynyl, and -Se-(CH2)m-R8, m
and R8
being defined above.
As used herein, the term "substituted" is contemplated to include all
permissible
substituents of organic compounds. In a broad aspect, the permissible
substituents
include acyclic and cyclic, branched and unbranched, carbocyclic and
heterocyclic,
aromatic and nonaromatic substituents of organic compounds. Illustrative
substituents
include, for example, those described herein above. The permissible
substituents can be
one or more and the same or different for appropriate organic compounds. For
purposes
of this invention, the heteroatoms such as nitrogen may have hydrogen
substituents
and/or any permissible substituents of organic compounds described herein
which satisfy
the valences of the heteroatoms. This invention is not intended to be limited
in any
manner by the permissible substituents of organic compounds.
It will be understood that "substitution" or "substituted with" includes the
implicit proviso that such substitution is in accordance with permitted
valence of the
substituted atom and the substituent, and that the substitution results in a
stable
compound, e.g., which does not spontaneously undergo transformation such as by
rearrangement, cyclization, elimination, etc.
The term "sulfamoyl" is art-recognized and includes a moiety that can be
represented by the general formula:
0
I I /Rio
-S-N
I I \ R
O 9
in which R9 and R10 are as defined above.
The term "sulfate" is art recognized and includes a moiety that can be
represented
by the general formula:
0
11
-O-S-ORai
O
in which R41 is as defined above.

CA 02386190 2002-04-11
WO 01/27135 PCT/US00/28479
-30-
The term "sulfonamido" is art recognized and includes a moiety that can be
represented by the general formula:
0
11
i S-R
R
9 O
in which R9 and R' 11 are as defined above.
The term "sulfonate" is art-recognized and includes a moiety that can be
represented by the general formula: :
0
I I
-S-OR41
0
in which R41 is an electron pair, hydrogen, alkyl, cycloalkyl, or aryl.
The terms "sulfoxido" or "sulfinyl", as used herein, refers to a moiety that
can be
represented by the general formula:
0
11
-S-R44
in which R44 is selected from the group consisting of hydrogen, alkyl,
alkenyl, alkynyl,
cycloalkyl, heterocyclyl, aralkyl, or aryl.
Analogous substitutions can be made to alkenyl and alkynyl groups to produce,
for example, aminoalkenyls, aminoalkynyls, amidoalkenyls, amidoalkynyls,
iminoalkenyls, iminoalkynyls, thioalkenyls, thioalkynyls, carbonyl-substituted
alkenyls
or alkynyls.
As used herein, the definition of each expression, e.g., alkyl, m, n, etc.,
when it
occurs more than once in any structure, is intended to be independent of its
definition
elsewhere in the same structure.
The terms triflyl, tosyl, mesyl, and nonaflyl are art-recognized and refer to
trifluoromethanesulfonyl, p-toluenesulfonyl, methanesulfonyl, and
nonafluorobutanesulfonyl groups, respectively. The terms triflate, tosylate,
mesylate, and

CA 02386190 2006-08-30
-31-
nonaflate are art-recognized and refer to trifluoromethanesulfonate ester, p-
toluenesulfonate ester, methanesulfonate ester, and nonafluorobutanesulfonate
ester
functional groups and molecules that contain said groups, respectively.
The abbreviations Me, Et, Ph, Tf, Nf, Ts, Ms represent methyl, ethyl, phenyl,
trifluoromethanesulfonyl, nonafluorobutanesulfonyl, p-toluenesulfonyl and
methanesulfonyl, respectively. A more comprehensive list of the abbreviations
utilized
by organic chemists of ordinary skill in the art appears in the first issue of
each volume
of the Journal of Organic Chemistry; this list is typically presented in a
table entitled
Standard List of Abbreviations.
Certain compounds of the present invention may exist in particular geometric
or
stereoisomeric forms. The present invention contemplates all such compounds,
including
cis- and trans-isomers, R- and S-enantiomers, diastereomers, (D)-isomers, (L)-
isomers,
the racemic mixtures thereof, and other mixtures thereof, as falling within
the scope of
the invention. Additional asymmetric carbon atoms may be present in a
substituent such
as an alkyl group. All such isomers, as well as mixtures thereof, are intended
to be
included in this invention.
If, for instance, a particular enantiomer of a compound of the present
invention is
desired, it may be prepared by asymmetric synthesis, or by derivation with a
chiral
auxiliary, where the resulting diastereomeric mixture is separated and the
auxiliary
group cleaved to provide the pure desired enantiomers. Altematively, where the
molecule contains a basic functional group, such as amino, or an acidic
functional group,
such as carboxyl, diastereomeric salts may be formed with an appropriate
optically
active acid or base, followed by resolution of the diastereomers thus formed
by
fractional crystallization or chromatographic means well known in the art, and
subsequent recovery of the pure enantiomers.
Contemplated equivalents of the compounds described above include compounds
which otherwise correspond thereto, and which have the same general properties
thereof
(e.g., the ability to inhibit hedgehog signaling), wherein one or more simple
variations of
substituents are made which do not adversely affect the efficacy of the
compound. In

CA 02386190 2002-04-11
WO 01/27135 PCT/US00/28479
-32-
general, the compounds of the present invention may be prepared by the methods
illustrated in the general reaction schemes as, for example, described below,
or by
modifications thereof, using readily available starting materials, reagents
and
conventional synthesis procedures. In these reactions, it is also possible to
make use of
variants which are in themselves known, but are not mentioned here.
For purposes of this invention, the chemical elements are identified in
accordance with the Periodic Table of the Elements, CAS version, Handbook of
Chemistry and Physics, 67th Ed., 1986-87, inside cover. Also for purposes of
this
invention, the term "hydrocarbon" is contemplated to include all permissible
compounds
having at least one hydrogen and one carbon atom. In a broad aspect, the
permissible
hydrocarbons include acyclic and cyclic, branched and unbranched, carbocyclic
and
heterocyclic, aromatic and nonaromatic organic compounds which can be
substituted or
unsubstituted.
III. Exemplary Compounds of the Invention.
As described in further detail below, it is contemplated that the subject
methods
can be carried out using any of a variety of different steroidal alkaloids
which can be
readily identified, e.g., by such drug screening assays as described herein.
Steroidal
alkaloids have a fairly complex nitrogen-containing nucleus. Two exemplary
classes of
steroidal alkaloids for use in the subject methods are the Solanum type and
the Veratrum
type. The above notwithstanding, in a preferred embodiment, the methods and
compositions of the present invention make use of compounds having a steroidal
alkaloid ring system of cyclopamine.
There are more than 50 naturally occuring veratrum alkaloids including
veratramine, cyclopamine, cycloposine, jervine, and muldamine occurring in
plants of
the Veratrum spp. The Zigadenus spp., death camas, also produces several
veratrum-type
of steroidal alkaloids including zygacine. In general, many of the veratrum
alkaloids
(e.g., jervine, cyclopamine and cycloposine) consist of a modified steroid
skeleton
attached spiro to a furanopiperidine. A typical veratrum-type alkaloid may be
represented by:

CA 02386190 2002-04-11
WO 01/27135 PCT/US00/28479
-33-
N N
R6 R6
R R
R4, O R4,
R3 R3
R2 "'C or R2 "'C \ \
An example of the Solanum type is solanidine. This steroidal alkaloid is the
nucleus (i.e., aglycone) for two important glycoalkaloids, solanine and
chaconine, found
in potatoes. Other plants in the Solanum family including various nightshades,
Jerusalem
cherries, and tomatoes also contain solanum-type glycoalkaloids.
Glycoalkaloids are
glycosides of alkaloids. A typical solanum-type alkaloid may be represented
by:
R20
R4
R5
N
R3 R21
R2
Based on these structures, and the possibility that certain unwanted side
effects
can be reduced by some manipulation of the structure, a wide range of
steroidal alkaloids
are contemplated as potential smoothened antagonists for use in the subject
method. For
example, compounds useful in the subject methods include steroidal alkaloids
represented in the general formulas (I), or unsaturated forms thereof and/or
seco-, nor- or
homo-derivatives thereof:
R6 R7
R6
R4 R 7 R4 R7
R5 or R 7
R2 R2 R5
R3 R3

CA 02386190 2002-04-11
WO 01/27135 PCT/US00/28479
-34-
Formula I
wherein, as valence and stability permit,
R2, R3, R4, and R5, represent one or more substitutions to the ring to which
each
is attached, for each occurrence, independently represent hydrogen, halogens,
alkyls,
alkenyls, alkynyls, aryls, hydroxyl, =0, =S, alkoxyl, silyloxy, amino, nitro,
thiol,
amines, imines, amides, phosphoryls, phosphonates, phosphines, carbonyls,
carboxyls,
carboxamides, anhydrides, silyls, ethers, thioethers, alkylsulfonyls,
arylsulfonyls,
selenoethers, ketones, aldehydes, esters, sugar (e.g., monosaccharide,
disaccharide,
polysaccharide, etc.), carbamate (e.g., attached to the steroid at oxygen),
carbonate, or -
(CH2)m-R8;
R6, R7, and R'7, are absent or represent, independently, halogens, alkyls,
alkenyls, alkynyls, aryls, hydroxyl, =0, =S, alkoxyl, silyloxy, amino, nitro,
thiol,
amines, imines, amides, phosphoryls, phosphonates, phosphines, carbonyls,
carboxyls,
carboxamides, anhydrides, silyls, ethers, thioethers, alkylsulfonyls,
arylsulfonyls,
selenoethers, ketones, aldehydes, esters, or -(CH2)m-R8, or
R6 and R7, or R7 and R'7, taken together form a ring or polycyclic ring, e.g.,
which is substituted or unsubstituted,
with the proviso that at least one of R6, R7, or R'7 is present and includes
an
amine, e.g., as one of the atoms which makes up the ring;
R8 represents an aryl, a cycloalkyl, a cycloalkenyl, a heterocycle, or a
polycycle;
and
m is an integer in the range 0 to 8 inclusive.
In certain embodiments, R2 represents =0, sugar (e.g., monosaccharide,
disaccharide, polysaccharide, etc.), carbamate (e.g., attached to the steroid
at oxygen),
ester (e.g., attached to the steroid at oxygen), carbonate, or alkoxy.
Substituents such as
carbamate, ester, carbonate, and alkoxy may be substituted or unsubstituted,
e.g., may

CA 02386190 2002-04-11
WO 01/27135 PCT/US00/28479
-35-
include additional functional groups such as aryl, aralkyl, heteroaryl,
heteroaralkyl,
amide, acylamino, carbonyl, ester, carbamate, urea, ketone, sulfonamide, etc.
In certain embodiments, the amine of R6, R,, or R'7 is a tertiary amine.
In particular embodiments, R3, for each occurrence, is an -OH, alkyl, -0-
alkyl, -
C(O)-alkyl, or -C(O)-R8.
In particular embodiments, R4, for each occurrence, is an absent, or
represents -
OH, =0, alkyl, -0-alkyl, -C(O)-alkyl, or -C(O)-R8.
In particular embodiments, two of R6, R7, and R'7 taken together form a
nitrogen-containing ring, such as a furanopiperidine, such as perhydrofuro[3,2-
b]pyridine, a pyranopiperidine, a quinoline, an indole, a pyranopyrrole, a
naphthyridine,
a thiofuranopiperidine, or a thiopyranopiperidine.
In certain embodiments, the nitrogen-containing ring comprises a tertiary
amine,
e.g., by having an extraannular substitutent on the nitrogen atom, e.g., an
alkyl
substituted with, for example, aryl, aralkyl, heteroaryl, heteroaralkyl,
amide, acylamino,
carbonyl, ester, carbamate, urea, ketone, sulfonamide, etc. In certain
embodiments, the
extraannular substituent of the tertiary amine is a hydrophobic substituent.
In certain
embodiments, the hydrophobic extraannular substituent includes an aryl,
heteroaryl,
carbocyclyl, heterocyclyl, or polycyclyl group, such as biotin, a zwitterionic
complex of
boron, a steroidal polycycle, etc. In certain embodiments, the hydrophobic
substituent
may consist essentially of a combination of alkyl, amido, acylamino, ketone,
ester, ether,
halogen, alkenyl, alkynyl, aryl, aralkyl, urea, or similar functional groups,
including
between 5 and 40 non-hydrogen atoms, more preferably between 5 and 20 non-
hydrogen
atoms.
In particular embodiments, R8 represents an aryl, a cycloalkyl, a
cycloalkenyl, a
heterocycle, or a polycycle, and preferably R8 is a piperidine, pyrrolidine,
pyridine,
pyrimidine, morpholine, thiomorpholine, pyridazine, etc.

CA 02386190 2002-04-11
WO 01/27135 PCT/US00/28479
-36-
In certain preferred embodiments, the definitions outlined above apply, and
the
subject compounds are represented by general formula Ia or unsaturated forms
thereof
and/or seco-, nor- or homo-derivatives thereof:
R6 R7
R6
R4 ""R' 7 R4
R7
t 7
R5 or PRS R
R2 _CJ R2 R3 R3
Formula Ia
In certain embodiments, the steroidal alkaloid is represented in the general
formula (II), or unsaturated fonms thereof and/or seco-, nor- or homo-
derivatives thereof:
R4 R6 R6 R7
R5 R7 R4 R5 R~
.
; 7
R3 7 or R3 ,
R2 5 R2 5
6 6
R6 R7
X R 7
R3
or
\
R2
Formula II
wherein R2, R3, R4, R5, R6, R7, and R'7 are as defined above, and X represents
O or S, though preferably O.
In certain embodiments, R2 represents =0, sugar (e.g., monosaccharide,
disaccharide, polysaccharide, etc.), carbamate (e.g., attached to the steroid
at oxygen),

CA 02386190 2002-04-11
WO 01/27135 PCT/US00/28479
-37-
ester (e.g., attached to the steroid at oxygen), carbonate, or alkoxy.
Substituents such as
carbamate, ester, carbonate, and alkoxy may be substituted or unsubstituted,
e.g., may
include additional functional groups such as aryl, aralkyl, heteroaryl,
heteroaralkyl,
amide, acylamino, carbonyl, ester, carbamate, urea, ketone, sulfonamide, etc.
In certain embodiments, the amine of R6, R71 or R'7 is a tertiary amine, e.g.,
substituted with a substituted or unsubstituted alkyl. In certain embodiments,
the amine
is part of a bicyclic ring system formed from R7 and R'7, e.g., a
furanopiperidine system,
and the third substitutent is an alkyl substituted with, for example, aryl,
aralkyl,
heteroaryl, heteroaralkyl, amide, acylamino, carbonyl, ester, carbamate, urea,
ketone,
sulfonamide, etc. In certain embodiments, the extraannular substituent of the
tertiary
amine is a hydrophobic substituent. In certain embodiments, the hydrophobic
extraannular substituent includes an aryl, heteroaryl, carbocyclyl,
heterocyclyl, or
polycyclyl group, such as biotin, a zwitterionic complex of boron, a steroidal
polycycle,
etc. In certain embodiments, the hydrophobic substituent may consist
essentially of a
combination of alkyl, amido, acylamino, ketone, ester, ether, halogen,
alkenyl, alkynyl,
aryl, aralkyl, urea, or similar functional groups, including between 5 and 40
non-
hydrogen atoms, more preferably between 5 and 20 non-hydrogen atoms.
In certain preferred embodiments, the definitions outlined above apply, and
the
subject compounds are represented by general formula IIa or unsaturated forms
thereof
and/or seco-, nor- or homo-derivatives thereof:
R4 R6 R6 R7
R5 R7 AR R5 R
I
; 7
~ ,
R3 R 7 or R2 O'e( 5 6 R2
6

CA 02386190 2002-04-11
WO 01/27135 PCT/US00/28479
-38-
R6 R7
X
7
R3
or
\
R2
Formula IIa
In certain embodiments, the steroidal alkaloid is represented in the general
formula (III), or unsaturated forms thereof and/or seco-, nor- or homo-
derivatives
thereof:
R4
R5 A R9 R4
or
dflR5mR9
R2 R2 6
Formula III
wherein
R2, R3, R4, R5 and R8 are as defined above;
A and B represent monocyclic or polycyclic groups;
T represents an alkyl, an aminoalkyl, a carboxyl, an ester, an amide, ether or
amine linkage of 1-10 bond lengths;
T' is absent, or represents an alkyl, an aminoalkyl, a carboxyl, an ester, an
amide,
ether or amine linkage of 1-3 bond lengths, wherein if T and T' are present
together, than
T and T' taken together with the ring A or B form a covalently closed ring of
5-8 ring
atoms;
R9 represents one or more substitutions to the ring A or B, which for each
occurrence, independently represent halogens, alkyls, alkenyls, alkynyls,
aryls, hydroxyl,

CA 02386190 2002-04-11
WO 01/27135 PCT/US00/28479
-39-
=0, =S, alkoxyl, silyloxy, amino, nitro, thiol, amines, imines, amides,
phosphoryls,
phosphonates, phosphines, carbonyls, carboxyls, carboxamides, anhydrides,
silyls,
ethers, thioethers, alkylsulfonyls, arylsulfonyls, selenoethers, ketones,
aldehydes, esters,
or -(CH2)m-R8; and
n and m are, independently, zero, 1 or 2;
with the proviso that A, or T, T', and B, taken together, include at least one
amine.
In certain embodiments, R2 represents =0, sugar (e.g., monosaccharide,
disaccharide, polysaccharide, etc.), carbamate (e.g., attached to the steroid
at oxygen),
ester (e.g., attached to the steroid at oxygen), carbonate, or alkoxy.
Substituents such as
carbamate, ester, carbonate, and alkoxy may be substituted or unsubstituted,
e.g., may
include additional functional groups such as aryl, aralkyl, heteroaryl,
heteroaralkyl,
amide, acylamino, carbonyl, ester, carbamate, urea, ketone, sulfonamide, etc.
In certain embodiments, the amine of A, or T, T', and B, is a tertiary amine,
e.g.,
substituted with a substituted or unsubstituted alkyl, e.g., substituted with
aryl, aralkyl,
heteroaryl, heteroaralkyl, amide, acylamino, carbonyl, ester, carbamate, urea,
ketone,
sulfonamide, etc. In certain embodiments, the extraannular substituent of the
tertiary
amine is a hydrophobic substituent. In certain embodiments, the hydrophobic
extraannular substituent includes an aryl, heteroaryl, carbocyclyl,
heterocyclyl, or
polycyclyl group, such as biotin, a zwitterionic complex of boron, a steroidal
polycycle,
etc. In certain embodiments, the hydrophobic substituent may consist
essentially of a
combination of alkyl, amido, acylamino, ketone, ester, ether, halogen,
alkenyl, alkynyl,
aryl, aralkyl, urea, or similar functional groups, including between 5 and 40
non-
hydrogen atoms, more preferably between 5 and 20 non-hydrogen atoms.
In certain preferred embodiments, the definitions outlined above apply, and
the
subject compounds are represented by general formula IIIa or unsaturated forms
thereof
and/or seco-, nor- or homo-derivatives thereof:

CA 02386190 2002-04-11
WO 01/27135 PCT/US00/28479
-40-
R4 R5 A R9 R4
R5 T
OR9
T
R3 n R3( '1.~
m or
R2 5
6 R2 5
6
Formula IIIa
For example, the subject methods can utilize smoothened antagonists based on
the veratrum-type steroidal alkaloids jervine, cyclopamine, cycloposine,
mukiamine or
veratramine, e.g., which may be represented in the general fonnula (IV), or
unsaturated
forms thereof and/or seco-, nor- or homo-derivatives thereof:
R22 N
R6 R22 N
R9 R6
R4 0 R9
R4
R5 or
R5
R2
R2
R3
R3
Formula IV
wherein
R2, R3, R4, R5, R6 and R9 are as defined above;
R22 is absent or represents an alkyl, an alkoxyl or -OH.
In certain embodiments, R2 represents =0, sugar (e.g., monosaccharide,
disaccharide, polysaccharide, etc.), carbamate (e.g., attached to the steroid
at oxygen),
ester (e.g., attached to the steroid at oxygen), carbonate, or alkoxy.
Substituents such as
carbamate, ester, carbonate, and alkoxy may be substituted or unsubstituted,
e.g., may
include additional functional groups such as aryl, aralkyl, heteroaryl,
heteroaralkyl,
amide, acylamino, carbonyl, ester, carbamate, urea, ketone, sulfonamide, etc.

CA 02386190 2002-04-11
WO 01/27135 PCT/US00/28479
-41-
In certain embodiments, R9 includes a substituent on nitrogen, e.g., a
substituted
or unsubstituted alkyl, e.g., substituted with, for example, aryl, aralkyl,
heteroaryl,
heteroaralkyl, amide, acylamino, carbonyl, ester, carbamate, urea, ketone,
sulfonamide,
etc. In certain embodiments, the extraannular substituent (e.g., R,) of the
tertiary amine
is a hydrophobic substituent. In certain embodiments, the hydrophobic
extraannular
substituent includes an aryl, heteroaryl, carbocyclyl, heterocyclyl, or
polycyclyl group,
such as biotin, a zwitterionic complex of boron, a steroidal polycycle, etc.
In certain
embodiments, the hydrophobic substituent may consist essentially of a
combination of
alkyl, amido, acylamino, ketone, ester, ether, halogen, alkenyl, alkynyl,
aryl, aralkyl,
urea, or similar functional groups, including between 5 and 40 non-hydrogen
atoms,
more preferably between 5 and 20 non-hydrogen atoms.
In certain preferred embodiments, the definitions outlined above apply, and
the
subject compounds are represented by general formula IVa or unsaturated forms
thereof
and/or seco-, nor- or homo-derivatives thereof:
R22
R6 R22 g N
R9 R6
R4 0 R9
R4
R5 or
R5
R2
R2
R3
R3
Formula IVa
In certain embodiments, the steroidal alkaloid is represented in the general
formula (V) or unsaturated forms thereof and/or seco-, nor- or homo-
derivatives thereof:

CA 02386190 2002-04-11
WO 01/27135 PCT/US00/28479
-42-
N N
R6 R6
R9 R9
R4, O R4,
R3 ,
or R3
R2 R2
R9
?%0
Ror
RR2
Formula V
wherein R2, R3, R4, R6 and Rq are as defined above;
In certain embodiments, R2 represents =0, sugar (e.g., monosaccharide,
disaccharide, polysaccharide, etc.), carbamate (e.g., attached to the steroid
at oxygen),
ester (e.g., attached to the steroid at oxygen), carbonate, or alkoxy.
Substituents such as
carbamate, ester, carbonate, and alkoxy may be substituted or unsubstituted,
e.g., may
include additional functional groups such as aryl, aralkyl, heteroaryl,
heteroaralkyl,
amide, acylamino, carbonyl, ester, carbamate, urea, ketone, sulfonamide, etc.
In certain embodiments, R9 includes a substituent on nitrogen, e.g., a
substituted
or unsubstituted alkyl, e.g., substituted with, for example, aryl, aralkyl,
heteroaryl,
heteroaralkyl, amide, acylamino, carbonyl, ester, carbamate, urea, ketone,
sulfonamide,
etc.
In certain embodiments, the extraannular substituent of the tertiary amine
(e.g.,
R9) is a hydrophobic substituent. In certain embodiments, the hydrophobic
extraannular
substituent includes an aryl, heteroaryl, carbocyclyl, heterocyclyl, or
polycyclyl group,

CA 02386190 2002-04-11
WO 01/27135 PCT/US00/28479
- 43 -
such as biotin, a zwitterionic complex of boron, a steroidal polycycle, etc.
In certain
embodiments, the hydrophobic substituent may consist essentially of a
combination of
alkyl, amido, acylamino, ketone, ester, ether, halogen, alkenyl, alkynyl,
aryl, aralkyl,
urea, or similar functional groups, including between 5 and 40 non-hydrogen
atoms,
more preferably between 5 and 20 non-hydrogen atoms.
In certain preferred embodiments, the definitions outlined above apply, and
the
subject compounds are represented by general formula Va or unsaturated forms
thereof
and/or seco-, nor- or homo-derivatives thereof:
N
R6 R6
R9 R9
R 3 4, O R4,
R
or
R2 R2 R3
\
N
R6
R9
R4, O
or
R3
R2
Formula Va
Another class of smoothened antagonists can be based on the veratrum-type
steroidal alkaloids resmebling verticine and zygacine, e.g., general formula
(VI), or
unsaturated forms thereof and/or seco-, nor- or homo-derivatives thereof:

CA 02386190 2002-04-11
WO 01/27135 PCT/US00/28479
-44-
R9
R9
N
R4 N
R9 R4
R9
or
R2 R5 ' _0 1 R2 1 R5
R3 0 R3
Formula VI
wherein R2, R3, R4, R5 and R9 are as defined above;
In certain embodiments, R2 represents =0, sugar (e.g., monosaccharide,
disaccharide, polysaccharide, etc.), carbamate (e.g., attached to the steroid
at oxygen),
ester (e.g., attached to the steroid at oxygen), carbonate, or alkoxy.
Substituents such as
carbamate, ester, carbonate, and alkoxy may be substituted or unsubstituted,
e.g., may
include additional functional groups such as aryl, aralkyl, heteroaryl,
heteroaralkyl,
amide, acylamino, carbonyl, ester, carbamate, urea, ketone, sulfonamide, etc.
In certain preferred embodiments, the definitions outlined above apply, and
the
subject compounds are represented by general formula VIa or unsaturated forms
thereof
and/or seco-, nor- or homo-derivatives thereof:

CA 02386190 2002-04-11
WO 01/27135 PCT/US00/28479
- 45 -
R9
R9
N
R4 N
R9 R4
R9
or
R2 R5
R2 R5
R3
0 R3
Formula VIa
In certain embodiments, the steroidal alkaloid is represented in the general
formula (VII) or unsaturated forms thereof and/or seco-, nor- or homo-
derivatives
thereof:
R9
R4
N R9
R2 R5
R3
Formula VII
wherein R2, R3, R4, R5 and Rq are as defined above.
In certain embodiments, R2 represents =0, sugar (e.g., monosaccharide,
disaccharide, polysaccharide, etc.), carbamate (e.g., attached to the steroid
at oxygen),
ester (e.g., attached to the steroid at oxygen), carbonate, or alkoxy.
Substituents such as
carbamate, ester, carbonate, and alkoxy may be substituted or unsubstituted,
e.g., may
include additional functional groups such as aryl, aralkyl, heteroaryl,
heteroaralkyl,
amide, acylamino, carbonyl, ester, carbamate, urea, ketone, sulfonamide, etc.

CA 02386190 2002-04-11
WO 01/27135 PCT/US00/28479
-46-
In certain preferred embodiments, the definitions outlined above apply, and
the
subject compounds are represented by general formula VIIa or unsaturated forms
thereof
and/or seco-, nor- or homo-derivatives thereof:
R9 H
R4
111'R9
N
R2 R5
R3
Formula VIIa
In certain embodiments, the subject antagonists and activators can be chosen
on
the basis of their selectively for the smoothened pathway. This selectivity
can be for the
smoothened pathway versus other steroid-mediated pathways (such as
testosterone or
estrogen mediated activities), as well as selectivity for particular
hedgehog/ptc/smoothened pathways, e.g., which isotype specific for ptc (e.g.,
ptc-1, ptc-
2) or hedgehog (e.g., Shh, Ihh, Dhh, etc). For instance, the subject method
may employ
steroidal alkaloids which do not substantially interfere with the biological
activity of
such steroids as aldosterone, androstane, androstene, androstenedione,
androsterone,
cholecalciferol, cholestane, cholic acid, corticosterone, cortisol, cortisol
acetate,
cortisone, cortisone acetate, deoxycorticosterone, digitoxigenin,
ergocalciferol,
ergosterol, estradiol-17-a, estradiol-17-(3, estriol, estrane, estrone,
hydrocortisone,
lanosterol, lithocholic acid, mestranol, P-methasone, prednisone, pregnane,
pregnenolone, progesterone, spironolactone, testosterone, triamcinolone and
their
derivatives, at least so far as those activities are unrelated to ptc related
signaling.
In one embodiment, the subject steroidal alkaloid for use in the present
method
has a kd for members of the nuclear hormone receptor superfamily of greater
than 1 M,
and more preferably greater than 1 mM, e.g., it does not bind estrogen,
testosterone

CA 02386190 2002-04-11
WO 01/27135 PCT/US00/28479
-47-
receptors or the like. Preferably, the subject smoothened antagonist has no
estrogenic
activity at physiological concentrations (e.g., in the range of 1 ng-1 mg/kg).
In this manner, untoward side effects which may be associated certain members
of the steroidal alkaloid class can be reduced. For example, using the drug
screening
assays described herein, the application of combinatorial and medicinal
chemistry
techniques to the steroidal alkaloids provides a means for reducing such
unwanted
negative side effects including personality changes, shortened life spans,
cardiovascular
diseases and vascular occlusion, organ toxicity, hyperglycemia and diabetes,
Cushnoid
features, "wasting" syndrome, steroidal glaucoma, hypertension, peptic ulcers,
and
increased susceptibility to infections. For certain embodiments, it will be
benefical to
reduce the teratogenic activity relative to jervine, as for example, in the
use of the
subject method to selectively inhibit spermatogenesis.
In a preferred embodiment, the subject antagonists are steroidal alkaloids
other
than spirosolane, tomatidine, jervine, etc.
In particular embodiments, the steroidal alkaloid is chosen for use because it
is
more selective for one patched isoform over the next, e.g., 10-fold, and more
preferably
at least 100- or even 1000-fold more selective for one patched pathway (ptc-1,
ptc-2)
over another. Likewise, the steroidal alkaloid may be chosen for use because
it is more
selective for one smoothened isoform over the next, e.g., 10-fold, and more
preferably at
least 100- or even 1000-fold more selective for one wild-type smoothened
protein
(should various isofonms exist) or for activated smoothened mutants relative
to wild-type
smoothened.In certain embodiments, the subject method can be carried out
conjointly
with the administration of growth and/or trophic factors, or compositions
which also act
on other parts of the hedgehog/smoothened pathway.. For instance, it is
contemplated
that the subject methods can include treatment with an agent that modulates
cAMP
levels, e.g., increasing or decreasing intracellular levels of cAMP.
In one embodiment, the subject method utilizes a smoothened antagonist, and
the
conjoint agent elevates cAMP levels in order to enhance the efficacy of the
smoothened
antagonist.

CA 02386190 2002-04-11
WO 01/27135 PCT/US00/28479
-48-
For example, compounds which may activate adenylate cyclase include forskolin
(FK), cholera toxin (CT), pertussis toxin (PT), prostaglandins (e.g., PGE-1
and PGE-2),
colforsin and (3-adrenergic receptor agonists. (3-Adrenergic receptor agonists
(sometimes
referred to herein as "(3-adrenergic agonists") include albuterol, bambuterol,
bitolterol,
carbuterol, clenbuterol, clorprenaline, denopamine, dioxethedrine, dopexamine,
ephedrine, epinephrine, etafedrine, ethylnorepinephrine, fenoterol,
formoterol,
hexoprenaline, ibopamine, isoetharine, isoproterenol, mabuterol,
metaproterenol,
methoxyphenamine, norepinephrine, oxyfedrine, pirbuterol, prenalterol,
procaterol,
propranolol, protokylol, quinterenol, reproterol, rimiterol, ritodrine,
salmefamol,
soterenol, salmeterol, terbutaline, tretoquinol, tulobuterol, and xamoterol.
Compounds which may inhibit a cAMP phosphodiesterase include amrinone,
milrinone, xanthine, methylxanthine, anagrelide, cilostamide, medorinone,
indolidan,
rolipram, 3 -isobutyl- 1 -methylxanthine (IBMX), chelerythrine, cilostazol,
glucocorticoids, griseolic acid, etazolate, caffeine, indomethacin, papverine,
MDL
12330A, SQ 22536, GDPssS, clonidine, type III and type IV phosphodiesterase
inhibitors, methylxanthines such as pentoxifylline, theophylline, theobromine,
pyrrolidinones and phenyl cycloalkane and cycloalkene derivatives (described
in PCT
publications Nos. WO 92/19594 and WO 92/10190), lisophylline, and fenoxamine.
Analogs of cAMP which may be useful in the present method include dibutyryl-
cAMP (db-cAMP), (8-(4)-chlorophenylthio)-cAMP (cpt-cAMP), 8-[(4-bromo-2,3-
dioxobutyl)thio]-cAMP, 2-[(4-bromo-2,3-dioxobutyl)thio]-cAMP, 8-bromo-cAMP,
dioctanoyl-cAMP, Sp-adenosine 3':5'-cyclic phosphorothioate, 8-piperidino-
cAMP, N6-
phenyl-cAMP, 8-methylamino-cAMP, 8-(6-aminohexyl)amino-cAMP, 2'-deoxy-cAMP,
N6,2'-O-dibutryl-cAMP, N6,2'-O-disuccinyl-cAMP, N6-monobutyryl-cAMP, 2'-O-
monobutyryl-cAMP, 2'-O-monobutryl-8-bromo-cAMP, N6-monobutryl-2'-deoxy-cAMP,
and 2'-O-monosuccinyl-cAMP.
Compounds which may reduce the levels or activity of cAMP include
prostaglandylinositol cyclic phosphate (cyclic PIP), endothelins (ET)-1 and -
3,
norepinepurine, K252a, dideoxyadenosine, dynorphins, melatonin, pertussis
toxin,
staurosporine, G; agonists, MDL 12330A, SQ 22536, GDPssS and clonidine, beta-

CA 02386190 2002-04-11
WO 01/27135 PCT/US00/28479
-49-
blockers, and ligands of G-protein coupled receptors. Additional compounds are
disclosed in U.S. Patent Nos. 5,891,875, 5,260,210, and 5,795,756.
Above-listed compounds useful in the subject methods may be modified to
increase the bioavailability, activity, or other pharmacologically relevant
property of the
compound. For example, forskolin has the formula:
Me
O
= Me
OH u/~I
O
OH Me
7
= 6
OAc
Me Me OH
Forskolin
Modifications of forskolin which have been found to increase the hydrophilic
character
of forskolin without severly attenuating the desired biological activity
include acylation
of the hydroxyls at C6 and/or C7 (after removal of the acetyl group) with
hydrophilic
acyl groups. In compounds wherein C6 is acylated with a hydrophilic acyl
group, C7
may optionally be deacetylated. Suitable hydrophilic acyl groups include
groups having
the structure -(CO)(CH2)oX, wherein X is OH or NR2; R is hydrogen, a C1-C4
alkyl
group, or two Rs taken together form a ring comprising 3-8 atoms, preferably 5-
7 atoms,
which may include heteroatoms (e.g., piperazine or morpholine rings); and n is
an
integer from 1-6, preferably from 1-4, even more preferably from 1-2. Other
suitable
hydrophilic acyl groups include hydrophilic amino acids or derivatives
thereof, such as
aspartic acid, glutamic acid, asparagine, glutamine, serine, threonine,
tyrosine, etc.,
including amino acids having a heterocyclic side chain. Forskolin, or other
compounds
listed above, modified by other possible hydrophilic acyl side chains known to
those of
skill in the art may be readily synthesized and tested for activity in the
present method.
Similarly, variants or derivatives of any of the above-listed compounds may be
effective as cAMP antagonists in the subject method, e.g., in order to
decrease cAMP
levels and potentiate the activity of a smoothened activator. Those skilled in
the art will
readily be able to synthesize and test such derivatives for suitable activity.

CA 02386190 2002-04-11
WO 01/27135 PCT/US00/28479
-50-
IV. Exemplary Applications of Method and Compositions
One aspect of the present invention relates to a method of modulating a
differentiated state, survival, and/or proliferation of a cell, such as a
normal cell or a cell
having a ptc loss-of-function, hedgehog gain-of-function, or smoothened gain-
of-
function, by contacting the cells with a compound as set forth above according
to the
subject method and as the circumstances may warrant.
For instance, it is contemplated by the invention that, in light of the
findings of
an apparently broad involvement of hedgehog, ptc, and smoothened in the
formation of
ordered spatial arrangements of differentiated tissues in vertebrates, the
subject method
could be used as part of a process for generating and/or maintaining an array
of different
vertebrate tissue both in vitro and in vivo. The compound, whether inductive
or anti-
inductive with respect to proliferation or differentiation of a given tissue,
can be, as
appropriate, any of the preparations described above.
For example, the present method of using subject compounds is applicable to
cell
culture techniques wherein it is desirable to control the proliferation or
differentiation of
the cell. A subject compound may be employed in a method directed towards
cells which
have a ptc loss-of-function, hedgehog gain-of-function, or smoothened gain-of-
function
phenotype. In vitro neuronal culture systems have proved to be fundamental and
indispensable tools for the study of neural development, as well as the
identification of
neurotrophic factors such as nerve growth factor (NGF), ciliary trophic
factors (CNTF),
and brain derived neurotrophic factor (BDNF). One use of the present method
may be in
cultures of neuronal stem cells, such as in the use of such cultures for the
generation of
new neurons and glia. In such embodiments of the subject method, the cultured
cells can
be contacted with a compound of the present invention in order to alter the
rate of
proliferation of neuronal stem cells in the culture and/or alter the rate of
differentiation,
or to maintain the integrity of a culture of certain terminally differentiated
neuronal cells.
In an exemplary embodiment, the subject method can be used to culture, for
example,
sensory neurons or, alternatively, motorneurons. Such neuronal cultures can be
used as
convenient assay systems as well as sources of implantable cells for
therapeutic
treatments.

CA 02386190 2002-04-11
WO 01/27135 PCT/US00/28479
-51 -
According to the present invention, large numbers of non-tumorigenic neural
progenitor cells can be perpetuated in vitro and their rate of proliferation
and/or
differentiation can be affected by contact with compounds of the present
invention.
Generally, a method is provided comprising the steps of isolating neural
progenitor cells
from an animal, perpetuating these cells in vitro or in vivo, preferably in
the presence of
growth factors, and regulating the differentiation of these cells into
particular neural
phenotypes, e.g., neurons and glia, by contacting the cells with a subject
compound.
Progenitor cells are thought to be under a tonic inhibitory influence which
maintains the progenitors in a suppressed state until their differentiation is
required.
However, recent techniques have been provided which permit these cells to be
proliferated, and unlike neurons which are terminally differentiated and
therefore non-
dividing, they can be produced in unlimited number and_ are highly suitable
for
transplantation into heterologous and autologous hosts with neurodegenerative
diseases.
By "progenitor" it is meant an oligopotent or multipotent stem cell which is
able
to divide without limit and, under specific conditions, can produce daughter
cells which
terminally differentiate such as into neurons and glia. These cells can be
used for
transplantation into a heterologous or autologous host. By heterologous is
meant a host
other than the animal from which the progenitor cells were originally derived.
By
autologous is meant the identical host from which the cells were originally
derived.
Cells can be obtained from embryonic, post-natal, juvenile or adult neural
tissue
from any animal. By any animal is meant any multicellular animal which
contains
nervous tissue. More particularly, is meant any fish, reptile, bird, amphibian
or mammal
and the like. The most preferable donors are mammals, especially mice and
humans.
In the case of a heterologous donor animal, the animal may be euthanized, and
the brain and specific area of interest removed using a sterile procedure.
Brain areas of
particular interest include any area from which progenitor cells can be
obtained which
will serve to restore function to a degenerated area of the host's brain.
These regions
include areas of the central nervous system (CNS) including the cerebral
cortex,
cerebellum, midbrain, brainstem, spinal cord and ventricular tissue, and areas
of the
peripheral nervous system (PNS) including the carotid body and the adrenal
medulla.

CA 02386190 2002-04-11
WO 01/27135 PCT/USOO/28479
-52-
More particularly, these areas include regions in the basal ganglia,
preferably the
striatum which consists of the caudate and putamen, or various cell groups
such as the
globus pallidus, the subthalamic nucleus, the nucleus basalis which is found
to be
degenerated in Alzheimer's Disease patients, or the substantia nigra pars
compacta which
is found to be degenerated in Parkinson's Disease patients.
Human heterologous neural progenitor cells may be derived from fetal tissue
obtained from elective abortion, or from a post-natal, juvenile or adult organ
donor.
Autologous neural tissue can be obtained by biopsy, or from patients
undergoing
neurosurgery in which neural tissue is removed, in particular during epilepsy
surgery,
and more particularly during temporal lobectomies and hippocampalectomies.
Cells can be obtained from donor tissue by dissociation of individual cells
from
the connecting extracellular matrix of the tissue. Dissociation can be
obtained using any
known procedure, including treatment with enzymes such as trypsin, collagenase
and the
like, or by using physical methods of dissociation such as with a blunt
instrument or by
mincing with a scalpel to a allow outgrowth of specific cell types from a
tissue.
Dissociation of fetal cells can be carried out in tissue culture medium, while
a preferable
medium for dissociation of juvenile and adult cells is artificial cerebral
spinal fluid
(aCSF). Regular aCSF contains 124 mM NaCI, 5 mM KCI, 1.3 mM MgC12, 2 mM
CaC12, 26 mM NaHCO3, and 10 mM D-glucose. Low Ca2+ aCSF contains the same
ingredients except for MgCl2 at a concentration of 3.2 mM and CaC12 at a
concentration
of 0.1 mM.
Dissociated cells can be placed into any known culture medium capable of
supporting cell growth, including MEM, DMEM, RPMI, F-12, and the like,
containing
supplements which are required for cellular metabolism such as glutamine and
other
amino acids, vitamins, minerals and useful proteins such as transferrin and
the like.
Medium may also contain antibiotics to prevent contamination with yeast,
bacteria and
fungi such as penicillin, streptomycin, gentamicin and the like. In some
cases, the
medium may contain serum derived from bovine, equine, chicken and the like. A
particularly preferable medium for cells is a mixture of DMEM and F-12.

CA 02386190 2002-04-11
WO 01/27135 PCT/US00/28479
-53-
Conditions for culturing should be close to physiological conditions. The pH
of
the culture media should be close to physiological pH, preferably between pH 6-
8, more
preferably close to pH 7, even more particularly about pH 7.4. Cells should be
cultured
at a temperature close to physiological temperature, preferably between 30 C-
40 C,
more preferably between 32 C-38 C, and most preferably between 35 C-37 C.
Cells can be grown in suspension or on a fixed substrate, but proliferation of
the
progenitors is preferably done in suspension to generate large numbers of
cells by
formation of "neurospheres" (see, for example, Reynolds et al. (1992) Science
255:1070-
1709; and PCT Publications W093/01275, W094/09 1 1 9, W094/10292, and
W094/16718). In the case of propagating (or splitting) suspension cells,
flasks are
shaken well and the neurospheres allowed to settle on the bottom corner of the
flask. The
spheres are then transferred to a 50 ml centrifuge tube and centrifuged at low
speed. The
medium is aspirated, the cells resuspended in a small amount of medium with
growth
factor, and the cells mechanically dissociated and resuspended in separate
aliquots of
media.
Cell suspensions in culture medium are supplemented with any growth factor
which allows for the proliferation of progenitor cells and seeded in any
receptacle
capable of sustaining cells, though as set out above, preferably in culture
flasks or roller
bottles. Cells typically proliferate within 3-4 days in a 37 C incubator, and
proliferation
can be reinitiated at any time after that by dissociation of the cells and
resuspension in
fresh medium containing growth factors.
In the absence of substrate, cells lift off the floor of the flask and
continue to
proliferate in suspension forming a hollow sphere of undifferentiated cells.
After
approximately 3-10 days in vitro, the proliferating clusters (neurospheres)
are fed every
2-7 days, and more particularly every 2-4 days by gentle centrifugation and
resuspension
in medium containing growth factor.
After 6-7 days in vitro, individual cells in the neurospheres can be separated
by
physical dissociation of the neurospheres with a blunt instrument, more
particularly by
triturating the neurospheres with a pipette. Single cells from the dissociated
neurospheres are suspended in culture medium containing growth factors, and

CA 02386190 2002-04-11
WO 01/27135 PCT/US00/28479
-54-
differentiation of the cells can be control in culture by plating (or
resuspending) the cells
in the presence of a subject compound.
To further illustrate other uses of the subject compounds, it is noted that
intracerebral grafting has emerged as an additional approach to central
nervous system
therapies. For example, one approach to repairing damaged brain tissues
involves the
transplantation of cells from fetal or neonatal animals into the adult brain
(Dunnett et al.
(1987) J Exp Biol 123:265-289; and Freund et al. (1985) J Neurosci 5:603-616).
Fetal
neurons from a variety of brain regions can be successfully incorporated into
the adult
brain, and such grafts can alleviate behavioral defects. For example, movement
disorder
induced by lesions of dopaminergic projections to the basal ganglia can be
prevented by
grafts of embryonic dopaminergic neurons. Complex cognitive functions that are
impaired after lesions of the neocortex can also be partially restored by
grafts of
embryonic cortical cells. The subject method can be used to regulate the
growth state in
the culture, or where fetal tissue is used, especially neuronal stem cells,
can be used to
regulate the rate of differentiation of the stem cells.
Stem cells useful in the present invention are generally known. For example,
several neural crest cells have been identified, some of which are multipotent
and likely
represent uncommitted neural crest cells, and others of which can generate
only one type
of cell, such as sensory neurons, and likely represent committed progenitor
cells. The
role of compounds employed in the present method to culture such stem cells
can be to
regulate differentiation of the uncommitted progenitor, or to regulate further
restriction
of the developmental fate of a committed progenitor cell towards becoming a
terminally
differentiated neuronal cell. For example, the present method can be used in
vitro to
regulate the differentiation of neural crest cells into glial cells, schwann
cells, chromaffin
cells, cholinergic sympathetic or parasympathetic neurons, as well as
peptidergic and
serotonergic neurons. The subject compounds can be used alone, or can be used
in
combination with other neurotrophic factors which act to more particularly
enhance a
particular differentiation fate of the neuronal progenitor cell.
In addition to the implantation of cells cultured in the presence of the
subject
compounds, yet another aspect of the present invention concerns the
therapeutic
application of a subject compound to regulate the growth state of neurons and
other

CA 02386190 2002-04-11
WO 01/27135 PCT/US00/28479
-55-
neuronal cells in both the central nervous system and the peripheral nervous
system. The
ability of ptc, hedgehog, and smoothened to regulate neuronal differentiation
during
development of the nervous system and also presumably in the adult state
indicates that,
in certain instances, the subject compounds can be expected to facilitate
control of adult
neurons with regard to maintenance, functional performance, and aging of
normal cells;
repair and regeneration processes in chemically or mechanically lesioned
cells; and
treatment of degeneration in certain pathological conditions. In light of this
understanding, the present invention specifically contemplates applications of
the subject
method to the treatment protocol of (prevention and/or reduction of the
severity of)
neurological conditions deriving from: (i) acute, subacute, or chronic injury
to the
nervous system, including traumatic injury, chemical injury, vascular injury
and deficits
(such as the ischemia resulting from stroke), together with
infectious/inflammatory and
tumor-induced injury; (ii) aging of the nervous system including Alzheimer's
disease;
(iii) chronic neurodegenerative diseases of the nervous system, including
Parkinson's
disease, Huntington's chorea, amylotrophic lateral sclerosis, diabetic
neuropathy, and the
like, as well as spinocerebellar degenerations; and (iv) chronic immunological
diseases
of the nervous system or affecting the nervous system, including multiple
sclerosis.
As appropriate, the subject method can also be used in generating nerve
prostheses for the repair of central and peripheral nerve damage. In
particular, where a
crushed or severed axon is intubulated by use of a prosthetic device, subject
compounds
can be added to the prosthetic device to regulate the rate of growth and
regeneration of
the dendritic processes. Exemplary nerve guidance channels are described in
U.S.
patents 5,092,871 and 4,955,892.
In another embodiment, the subject method can be used in the treatment of
neoplastic or hyperplastic transformations such as may occur in the central
nervous
system. For instance, the subject compounds can be utilized to cause such
transformed
cells to become either post-mitotic or apoptotic. The present method may,
therefore, be
used as part of a treatment for, e.g., malignant gliomas, meningiomas,
medulloblastomas, neuroectodermal tumors, and ependymomas.

CA 02386190 2002-04-11
WO 01/27135 PCT/US00/28479
-56-
In a preferred embodiment, the subject method can be used as part of a
treatment
regimen for malignant medulloblastoma and other primary CNS malignant
neuroectodermal tumors.
In certain embodiments, the subject method is used as part of treatment
program
for medulloblastoma. Medulloblastoma, a primary brain tumor, is the most
common
brain tumor in children. A medulloblastoma is a primitive neuroectodermal
tumor
arising in the posterior fossa. They account for approximately 25% of all
pediatric brain
tumors (Miller). Histologically, they are small round cell tumors commonly
arranged in
true rosettes, but may display some differentiation to astrocytes, ependymal
cells or
neurons (Rorke; Kleihues). PNET's may arise in other areas of the brain
including the
pineal gland (pineoblastoma) and cerebrum. Patients with tumors arising in the
supratentorial region generally fare worse than their PF counterparts.
MedulloblastomalPNET's are known to recur anywhere in the CNS after
resection, and can even metastasize to bone. Pretreatment evaluation should
therefore
include an examination of the spinal cord to exclude the possibility of
"dropped
metastases". Gadolinium-enhanced MRI has largely replaced myelography for this
purpose, and CSF cytology is obtained postoperatively as a routine procedure.
In other embodiments, the subject method is used as part of treatment program
for ependymomas. Ependymomas account for approximately 10% of the pediatric
brain
tumors in children. Grossly, they are tumors that arise from the ependymal
lining of the
ventricles and microscopically fonn rosettes, canals, and perivascular
rosettes. In the
CHOP series of 51 children reported with ependymomas, 1/4 were histologically
benign.
Approximately 2/3 arose from the region of the 4th ventricle. One third
presented in the
supratentorial region. Age at presentation peaks between birth and 4 years, as
demonstrated by SEER data as well as data from CHOP. The median age is about 5
years. Because so many children with this disease are babies, they often
require
multimodal therapy.
Yet another aspect of the present invention concerns the observation in the
art
that ptc, hedgehog, and/or smoothened are involved in morphogenic signals
involved in
other vertebrate organogenic pathways in addition to neuronal differentiation
as

CA 02386190 2002-04-11
WO 01/27135 PCT/US00/28479
-57-
described above, having apparent roles in other endodermal patterning, as well
as both
mesodermal and endodermal differentiation processes. Thus, it is contemplated
by the
invention that compositions comprising one or more of the subject compounds
can also
be utilized for both cell culture and therapeutic methods involving generation
and
maintenance of non-neuronal tissue.
In one embodiment, the present invention makes use of the discovery that ptc,
hedgehog, and smoothened are apparently involved in controlling the
development of
stem cells responsible for formation of the digestive tract, liver, lungs, and
other organs
which derive from the primitive gut. Shh serves as an inductive signal from
the
endoderm to the mesoderm, which is critical to gut morphogenesis. Therefore,
for
example, compounds of the instant method can be employed for regulating the
development and maintenance of an artificial liver which can have multiple
metabolic
functions of a normal liver. In an exemplary embodiment, the subject method
can be
used to regulate the proliferation and differentiation of digestive tube stem
cells to form
hepatocyte cultures which can be used to populate extracellular matrices, or
which can
be encapsulated in biocompatible polymers, to form both implantable and
extracorporeal
artificial livers.
In another embodiment, therapeutic compositions of subject compounds can be
utilized in conjunction with transplantation of such artificial livers, as
well as embryonic
liver structures, to regulate uptake of intraperitoneal implantation,
vascularization, and in
vivo differentiation and maintenance of the engrafted liver tissue.
In yet another embodiment, the subject method can be employed therapeutically
to regulate such organs after physical, chemical or pathological insult. For
instance,
therapeutic compositions comprising subject compounds can be utilized in liver
repair
subsequent to a partial hepatectomy.
The generation of the pancreas and small intestine from the embryonic gut
depends on intercellular signalling between the endodermal and mesodermal
cells of the
gut. In particular, the differentiation of intestinal mesoderm into smooth
muscle has been
suggested to depend on signals from adjacent endodermal cells. One candidate
mediator
of endodermally derived signals in the embryonic hindgut is Sonic hedgehog.
See, for

CA 02386190 2002-04-11
WO 01/27135 PCT/US00/28479
-58-
example, Apelqvist et al. (1997) Curr Biol 7:801-4. The Shh gene is expressed
throughout the embryonic gut endoderm with the exception of the pancreatic bud
endoderm, which instead expresses high levels of the homeodomain protein
Ipfl/Pdxl
(insulin promoter factor 1/pancreatic and duodenal homeobox 1), an essential
regulator
of early pancreatic development. Apelqvist et al., supra, have examined
whether the
differential expression of Shh in the embryonic gut tube controls the
differentiation of
the surrounding mesoderm into specialised mesoderm derivatives of the small
intestine
and pancreas. To test this, they used the promoter of the Ipfl/Pdxl gene to
selectively
express Shh in the developing pancreatic epithelium. In Ipfl/Pdxl- Shh
transgenic mice,
the pancreatic mesoderm developed into smooth muscle and interstitial cells of
Cajal,
characteristic of the intestine, rather than into pancreatic mesenchyme and
spleen. Also,
pancreatic explants exposed to Shh underwent a similar program of intestinal
differentiation. These results provide evidence that the - differential
expression of
endodermally derived Shh controls the fate of adjacent mesoderm at different
regions of
the gut tube.
In the context of the present invention, it is contemplated therefore that the
subject compounds can be used to control or regulate the proliferation and/or
differentiation of pancreatic tissue both in vivo and in vitro.
There are a wide variety of pathological cell proliferative and
differentiative
conditions for which the inhibitors of the present invention may provide
therapeutic
benefits, with the general strategy being, for example, the correction of
aberrant insulin
expression, or modulation of differentiation. More generally, however, the
present
invention relates to a method of inducing and/or maintaining a differentiated
state,
enhancing survival and/or affecting proliferation of pancreatic cells, by
contacting the
cells with the subject inhibitors. For instance, it is contemplated by the
invention that, in
light of the apparent involvement of ptc, hedgehog, and smoothened in the
formation of
ordered spatial arrangements of pancreatic tissues, the subject method could
be used as
part of a technique to generate and/or maintain such tissue both in vitro and
in vivo. For
instance, modulation of the function of hedgehog can be employed in both cell
culture
and therapeutic methods involving generation and maintenance P-cells and
possibly also
for non-pancreatic tissue, such as in controlling the development and
maintenance of

CA 02386190 2002-04-11
WO 01/27135 PCT/US00/28479
-59-
tissue from the digestive tract, spleen, lungs, urogenital organs (e.g.,
bladder), and other
organs which derive from the primitive gut.
In an exemplary embodiment, the present method can be used in the treatment of
hyperplastic and neoplastic disorders affecting pancreatic tissue,
particularly those
characterized by aberrant proliferation of pancreatic cells. For instance,
pancreatic
cancers are marked by abnormal proliferation of pancreatic cells which can
result in
alterations of insulin secretory capacity of the pancreas. For instance,
certain pancreatic
hyperplasias, such as pancreatic carcinomas, can result in hypoinsulinemia due
to
dysfunction of 0-cells or decreased islet cell mass. To the extent that
aberrant ptc,
hedgehog, and smoothened signaling may be indicated in disease progression,
the
subject regulators can be used to enhance regeneration of the tissue after
anti-tumor
therapy.
Moreover, manipulation of hedgehog signaling properties at different points
may
be useful as part of a strategy for reshaping/repairing pancreatic tissue both
in vivo and
in vitro. In one embodiment, the present invention makes use of the apparent
involvement of ptc, hedgehog, and smoothened in regulating the development of
pancreatic tissue. In general, the subject method can be employed
therapeutically to
regulate the pancreas after physical, chemical or pathological insult. In yet
another
embodiment, the subject method can be applied to to cell culture techniques,
and in
particular, may be employed to enhance the initial generation of prosthetic
pancreatic
tissue devices. Manipulation of proliferation and differentiation of
pancreatic tissue, for
example, by altering hedgehog activity, can provide a means for more carefully
controlling the characteristics of a cultured tissue. In an exemplary
embodiment, the
subject method can be used to augment production of prosthetic devices which
require
(3-islet cells, such as may be used in the encapsulation devices described in,
for example,
the Aebischer et al. U.S. Patent No. 4,892,538, the Aebischer et al. U.S.
Patent No.
5,106,627, the Lim U.S. Patent No. 4,391,909, and the Sefton U.S. Patent No.
4,353,888. Early progenitor cells to the pancreatic islets are multipotential,
and
apparently coactivate all the islet-specific genes from the time they first
appear. As
development proceeds, expression of islet-specific hormones, such as insulin,
becomes
restricted to the pattern of expression characteristic of mature islet cells.
The phenotype

CA 02386190 2002-04-11
WO 01/27135 PCT/US00/28479
-60-
of mature islet cells, however, is not stable in culture, as reappearence of
embryonal
traits in mature P-cells can be observed. By utilizing the subject compounds,
the
differentiation path or proliferative index of the cells can be regulated.
Furthermore, manipulation of the differentiative state of pancreatic tissue
can be
utilized in conjunction with transplantation of artificial pancreas so as to
promote
implantation, vascularization, and in vivo differentiation and maintenance of
the
engrafted tissue. For instance, manipulation of hedgehog function to affect
tissue
differentiation can be utilized as a means of maintaining graft viability.
Bellusci et al. (1997) Development 124:53 report that Sonic hedgehog regulates
lung mesenchymal cell proliferation in vivo. Accordingly, the present method
can be
used to regulate regeneration of lung tissue, e.g., in the treatment of
emphysema.
Fujita et al. (1997) Biochem Biophys Res Commun 238:658 reported that Sonic
hedgehog is expressed in human lung squamous carcinoma and adenocarcinoma
cells.
The expression of Sonic hedgehog was also detected in the human lung squamous
carcinoma tissues, but not in the normal lung tissue of the same patient. They
also
observed that Sonic hedgehog stimulates the incorporation of BrdU into the
carcinoma
cells and stimulates their cell growth, while anti-Shh-N inhibited their cell
growth. These
results suggest that a ptc, hedgehog, and/or smoothened is involved in the
cell growth of
such transformed lung tissue and therefore indicates that the subject method
can be used
as part of a treatment of lung carcinoma and adenocarcinomas, and other
proliferative
disorders involving the lung epithelia.
Many other tumors may, based on evidence such as involvement of the hedgehog
pathway in these tumors, or detected expression of hedgehog or its receptor in
these
tissues during development, be affected by treatment with the subject
compounds. Such
tumors include, but are by no means limited to, tumors related to Gorlin's
syndrome
(e.g., basal cell carcinoma, medulloblastoma, meningioma, etc.), tumors
evidenced in pct
knock-out mice (e.g., hemangioma, rhabdomyosarcoma, etc.), tumors resulting
from gli-
1 amplification (e.g., glioblastoma, sarcoma, etc.), tumors connected with
TRC8, a ptc
homolog (e.g., renal carcinoma, thyroid carcinoma, etc.), Ext-l-related tumors
(e.g.,
bone cancer, etc.), Shh-induced tumors (e.g., lung cancer, chondrosarcomas,
etc.), and

CA 02386190 2002-04-11
WO 01/27135 PCT/US00/28479
-61-
other tumors (e.g., breast cancer, urogenital cancer (e.g., kidney, bladder,
ureter,
prostate, etc.), adrenal cancer, gastrointestinal cancer (e.g., stomach,
intestine, etc.), etc.).
In still another embodiment of the present invention, compositions comprising
one or more of the subject compounds can be used in the in vitro generation of
skeletal
tissue, such as from skeletogenic stem cells, as well as the in vivo treatment
of skeletal
tissue deficiencies. The present invention particularly contemplates the use
of subject
compounds to regulate the rate of chondrogenesis and/or osteogenesis. By
"skeletal
tissue deficiency", it is meant a deficiency in bone or other skeletal
connective tissue at
any site where it is desired to restore the bone or connective tissue, no
matter how the
deficiency originated, e.g. whether as a result of surgical intervention,
removal of tumor,
ulceration, implant, fracture, or other traumatic or degenerative conditions.
For instance, the method of the present invention can be used as part of a
regimen for restoring cartilage function to a connective tissue. Such methods
are useful
in, for example, the repair of defects or lesions in cartilage tissue which is
the result of
degenerative wear such as that which results in arthritis, as well as other
mechanical
derangements which may be caused by trauma to the tissue, such as a
displacement of
torn meniscus tissue, meniscectomy, a laxation of a joint by a torn ligament,
malignment
of joints, bone fracture, or by hereditary disease. The present reparative
method is also
useful for remodeling cartilage matrix, such as in plastic or reconstructive
surgery, as
well as periodontal surgery. The present method may also be applied to
improving a
previous reparative procedure, for example, following surgical repair of a
meniscus,
ligament, or cartilage. Furthermore, it may prevent the onset or exacerbation
of
degenerative disease if applied early enough after trauma.
In one embodiment of the present invention, the subject method comprises
treating the afflicted connective tissue with a therapeutically sufficient
amount of a
subject compound to regulate a cartilage repair response in the connective
tissue by
managing the rate of differentiation and/or proliferation of chondrocytes
embedded in
the tissue. Such connective tissues as articular cartilage, interarticular
cartilage
(menisci), costal cartilage (connecting the true ribs and the sternum),
ligaments, and
tendons are particularly amenable to treatment in reconstructive and/or
regenerative
therapies using the subject method. As used herein, regenerative therapies
include

CA 02386190 2002-04-11
WO 01/27135 PCT/US00/28479
-62-
treatment of degenerative states which have progressed to the point of which
impairment
of the tissue is obviously manifest, as well as preventive treatments of
tissue where
degeneration is in its earliest stages or imminent.
In an illustrative embodiment, the subject method can be used as part of a
therapeutic intervention in the treatment of cartilage of a diarthroidal
joint, such as a
knee, an ankle, an elbow, a hip, a wrist, a knuckle of either a finger or toe,
or a
tempomandibular joint. The treatment can be directed to the meniscus of the
joint, to the
articular cartilage of the joint, or both. To further illustrate, the subject
method can be
used to treat a degenerative disorder of a knee, such as which might be the
result of
traumatic injury (e.g., a sports injury or excessive wear) or osteoarthritis.
The subject
regulators may be administered as an injection into the joint with, for
instance, an
arthroscopic needle. In some instances, the injected agent can be in the form
of a
hydrogel or other slow release vehicle in order to permit a more extended and
regular
contact of the agent with the treated tissue.
The present invention further contemplates the use of the subject method in
the
field of cartilage transplantation and prosthetic device therapies. However,
problems
arise, for instance, because the characteristics of cartilage and
fibrocartilage varies
between different tissue: such as between articular, meniscal cartilage,
ligaments, and
tendons, between the two ends of the same ligament or tendon, and between the
superficial and deep parts of the tissue. The zonal arrangement of these
tissues may
reflect a gradual change in mechanical properties, and failure occurs when
implanted
tissue, which has not differentiated under those conditions, lacks the ability
to
appropriately respond. For instance, when meniscal cartilage is used to repair
anterior
cruciate ligaments, the tissue undergoes a metaplasia to pure fibrous tissue.
By
regulating the rate of chondrogenesis, the subject method can be used to
particularly
address this problem, by helping to adaptively control the implanted cells in
the new
environment and effectively resemble hypertrophic chondrocytes of an earlier
developmental stage of the tissue.
In similar fashion, the subject method can be applied to enhancing both the
generation of prosthetic cartilage devices and to their implantation. The need
for
improved treatment has motivated research aimed at creating new cartilage that
is based

CA 02386190 2002-04-11
WO 01/27135 PCT/US00/28479
-63-
on collagen-glycosaminoglycan templates (Stone et al. (1990) Clin Orthop Relat
Red
252:129), isolated chondrocytes (Grande et al. (1989) J Orthop Res 7:208; and
Takigawa
et al. (1987) Bone Miner 2:449), and chondrocytes attached to natural or
synthetic
polymers (Walitani et al. (1989) J Bone Jt Surg 71B:74; Vacanti et al. (1991)
Plast
Reconstr Surg 88:753; von Schroeder et al. (1991) JBiomed Mater Res 25:329;
Freed et
al. (1993) JBiomed Mater Res 27:11; and the Vacanti et al. U.S. Patent No.
5,041,138).
For example, chondrocytes can be grown in culture on biodegradable,
biocompatible
highly porous scaffolds formed from polymers such as polyglycolic acid,
polylactic acid,
agarose gel, or other polymers which degrade over time as function of
hydrolysis of the
polymer backbone into innocuous monomers. The matrices are designed to allow
adequate nutrient and gas exchange to the cells until engraftment occurs. The
cells can
be cultured in vitro until adequate cell volume and density has developed for
the cells to
be implanted. One advantage of the matrices is that they can be cast or molded
into a
desired shape on an individual basis, so that the final product closely
resembles the
patient's own ear or nose (by way of example), or flexible matrices can be
used which
allow for manipulation at the time of implantation, as in a joint.
In one embodiment of the subject method, the implants are contacted with a
subject compound during certain stages of the culturing process in order to
manage the
rate of differentiation of chondrocytes and the formation of hypertrophic
chrondrocytes
in the culture.
In another embodiment, the implanted device is treated with a subject compound
in order to actively remodel the implanted matrix and to make it more suitable
for its
intended function. As set out above with respect to tissue transplants, the
artificial
transplants suffer from the same deficiency of not being derived in a setting
which is
comparable to the actual mechanical environment in which the matrix is
implanted. The
ability to regulate the chondrocytes in the matrix by the subject method can
allow the
implant to acquire characteristics similar to the tissue for which it is
intended to replace.
In yet another embodiment, the subject method is used to enhance attachment of
prosthetic devices. To illustrate, the subject method can be used in the
implantation of a
periodontal prosthesis, wherein the treatment of the surrounding connective
tissue
stimulates formation of periodontal ligament about the prosthesis.

CA 02386190 2002-04-11
WO 01/27135 PCT/US00/28479
-64-
In still further embodiments, the subject method can be employed as part of a
regimen for the generation of bone (osteogenesis) at a site in the animal
where such
skeletal tissue is deficient. Indian hedgehog is particularly associated with
the
hypertrophic chondrocytes that are ultimately replaced by osteoblasts. For
instance,
administration of a compound of the present invention can be employed as part
of a
method for regulating the rate of bone loss in a subject. For example,
preparations
comprising subject compounds can be employed, for example, to control
endochondral
ossification in the formation of a "model" for ossification.
In yet another embodiment of the present invention, a subject compound can be
used to regulate spermatogenesis. The hedgehog proteins, particularly Dhh,
have been
shown to be involved in the differentiation and/or proliferation and
maintenance of
testicular germ cells. Dhh expression is initiated in Sertoli cell precursors
shortly after
the activation of Sry (testicular determining gene) and persists in the testis
into the adult.
Males are viable but infertile, owing to a complete absence of mature sperm.
Examination of the developing testis in different genetic backgrounds suggests
that Dhh
regulates both early and late stages of spermatogenesis. Bitgood et al. (1996)
Curr Biol
6:298. In a preferred embodiment, the subject compound can be used as a
contraceptive.
In similar fashion, compounds of the subject method are potentially useful for
modulating normal ovarian function.
The subject method also has wide applicability to the treatment or prophylaxis
of
disorders afflicting epithelial tissue, as well as in cosmetic uses. In
general, the method
can be characterized as including a step of administering to an animal an
amount of a
subject compound effective to alter the growth state of a treated epithelial
tissue. The
mode of administration and dosage regimens will vary depending on the
epithelial
tissue(s) which is to be treated. For example, topical formulations will be
preferred
where the treated tissue is epidermal tissue, such as dermal or mucosal
tissues.
A method which "promotes the healing of a wound" results in the wound healing
more quickly as a result of the treatment than a similar wound heals in the
absence of the
treatment. "Promotion of wound healing" can also mean that the method
regulates the
proliferation and/or growth of, inter alia, keratinocytes, or that the wound
heals with less
scarring, less wound contraction, less collagen deposition and more
superficial surface

CA 02386190 2002-04-11
WO 01/27135 PCT/US00/28479
-65-
area. In certain instances, "promotion of wound healing" can also mean that
certain
methods of wound healing have improved success rates, (e.g., the take rates of
skin
grafts) when used together with the method of the present invention.
Despite significant progress in reconstructive surgical techniques, scarring
can be
an important obstacle in regaining normal function and appearance of healed
skin. This
is particularly true when pathologic scarring such as keloids or hypertrophic
scars of the
hands or face causes functional disability or physical deformity. In the
severest
circumstances, such scarring may precipitate psychosocial distress and a life
of
economic deprivation. Wound repair includes the stages of hemostasis,
inflammation,
proliferation, and remodeling. The proliferative stage involves multiplication
of
fibroblasts and endothelial and epithelial cells. Through the use of the
subject method,
the rate of proliferation of epithelial cells in and proximal to the wound can
be controlled
in order to accelerate closure of the wound and/or minimize the fonmation of
scar tissue.
The present treatment can also be effective as part of a therapeutic regimen
for
treating oral and paraoral ulcers, e.g., resulting from radiation and/or
chemotherapy.
Such ulcers commonly develop within days after chemotherapy or radiation
therapy.
These ulcers usually begin as small, painful irregularly shaped lesions
usually covered
by a delicate gray necrotic membrane and surrounded by inflammatory tissue. In
many
instances, lack of treatment results in proliferation of tissue around the
periphery of the
lesion on an inflammatory basis. For instance, the epithelium bordering the
ulcer usually
demonstrates proliferative activity, resulting in loss of continuity of
surface epithelium.
These lesions, because of their size and loss of epithelial integrity, dispose
the body to
potential secondary infection. Routine ingestion of food and water becomes a
very
painful event and, if the ulcers proliferate throughout the alimentary canal,
diarrhea
usually is evident with all its complicating factors. According to the present
invention, a
treatment for such ulcers which includes application of a subject compound can
reduce
the abnormal proliferation and differentiation of the affected epithelium,
helping to
reduce the severity of subsequent inflammatory events.
The subject method and compositions can also be used to treat wounds resulting
from denmatological diseases, such as lesions resulting from autoimmune
disorders such
as psoriasis. Atopic dermititis refers to skin trauma resulting from allergies
associated

CA 02386190 2002-04-11
WO 01/27135 PCT/USOO/28479
-66-
with an immune response caused by allergens such as pollens, foods, dander,
insect
venoms and plant toxins.
In other embodiments, antiproliferative preparations of subject compounds can
be used to inhibit lens epithelial cell proliferation to prevent post-
operative
complications of extracapsular cataract extraction. Cataract is an intractable
eye disease
and various studies on a treatment of cataract have been made. But at present,
the
treatment of cataract is attained by surgical operations. Cataract surgery has
been applied
for a long time and various operative methods have been examined.
Extracapsular lens
extraction has become the method of choice for removing cataracts. The major
medical
advantages of this technique over intracapsular extraction are lower incidence
of aphakic
cystoid macular edema and retinal detachment. Extracapsular extraction is also
required
for implantation of posterior chamber type intraocular lenses which are now
considered
to be the lenses of choice in most cases.
However, a disadvantage of extracapsular cataract extraction is the high
incidence of posterior lens capsule opacification, often called after-
cataract, which can
occur in up to 50% of cases within three years after surgery. After-cataract
is caused by
proliferation of equatorial and anterior capsule lens epithelial cells which
remain after
extracapsular lens extraction. These cells proliferate to cause Sommerling
rings, and
along with fibroblasts which also deposit and occur on the posterior capsule,
cause
opacification of the posterior capsule, which interferes with vision.
Prevention of after-
cataract would be preferable to treatment. To inhibit secondary cataract
formation, the
subject method provides a means for inhibiting proliferation of the remaining
lens
epithelial cells. For example, such cells can be induced to remain quiescent
by instilling
a solution containing a preparation of a subject compound into the anterior
chamber of
the eye after lens removal. Furthermore, the solution can be osmotically
balanced to
provide minimal effective dosage when instilled into the anterior chamber of
the eye,
thereby inhibiting subcapsular epithelial growth with some specificity.
The subject method can also be used in the treatment of corneopathies marked
by
comeal epithelial cell proliferation, as for example in ocular epithelial
disorders such as
epithelial downgrowth or squamous cell carcinomas of the ocular surface.

CA 02386190 2002-04-11
WO 01/27135 PCT/US00/28479
-67-
Levine et al. (1997) J Neurosci 17:6277 show that hedgehog proteins can
regulate mitogenesis and photoreceptor differentiation in the vertebrate
retina, and Ihh is
a candidate factor from the pigmented epithelium to promote retinal progenitor
proliferation and photoreceptor differentiation. Likewise, Jensen et al.
(1997)
Development 124:363 demonstrated that treatment of cultures of perinatal mouse
retinal
cells with the amino-terminal fragment of Sonic hedgehog results in an
increase in the
proportion of cells that incorporate bromodeoxuridine, in total cell numbers,
and in rod
photoreceptors, amacrine cells and Muller glial cells, suggesting that Sonic
hedgehog
promotes the proliferation of retinal precursor cells. Thus, the subject
method can be
used in the treatment of proliferative diseases of retinal cells and regulate
photoreceptor
differentiation.
Yet another aspect of the present invention relates to the use of the subject
method to control hair growth. Hair is basically composed of keratin, a tough
and
insoluble protein; its chief strength lies in its disulphide bond of cystine.
Each individual
hair comprises a cylindrical shaft and a root, and is contained in a follicle,
a flask-like
depression in the skin. The bottom of the follicle contains a finger-like
projection termed
the papilla, which consists of connective tissue from which hair grows, and
through
which blood vessels supply the cells with nourishment. The shaft is the part
that extends
outwards from the skin surface, whilst the root has been described as the
buried part of
the hair. The base of the root expands into the hair bulb, which rests upon
the papilla.
Cells from which the hair is produced grow in the bulb of the follicle; they
are extruded
in the form of fibers as the cells proliferate in the follicle. Hair "growth"
refers to the
formation and elongation of the hair fiber by the dividing cells.
As is well known in the art, the common hair cycle is divided into three
stages:
anagen, catagen and telogen. During the active phase (anagen), the epidermal
stem cells
of the dermal papilla divide rapidly. Daughter cells move upward and
differentiate to
form the concentric layers of the hair itself. The transitional stage,
catagen, is marked by
the cessation of mitosis of the stem cells in the follicle. The resting stage
is known as
telogen, where the hair is retained within the scalp for several weeks before
an emerging
new hair developing below it dislodges the telogen-phase shaft from its
follicle. From
this model it has become clear that the larger the pool of dividing stem cells
that

CA 02386190 2002-04-11
WO 01/27135 PCT/US00/28479
-68-
differentiate into hair cells, the more hair growth occurs. Accordingly,
methods for
increasing or reducing hair growth can be carried out by potentiating or
inhibiting,
respectively, the proliferation of these stem cells.
In certain embodiments, the subject method can be employed as a way of
reducing the growth of human hair as opposed to its conventional removal by
cutting,
shaving, or depilation. For instance, the present method can be used in the
treatment of
trichosis characterized by abnormally rapid or dense growth of hair, e.g.
hypertrichosis.
In an exemplary embodiment, subject compounds can be used to manage hirsutism,
a
disorder marked by abnormal hairiness. The subject method can also provide a
process
for extending the duration of depilation.
Moreover, because a subject compound will often be cytostatic to epithelial
cells,
rather than cytotoxic, such agents can be used to protect hair follicle cells
from cytotoxic
agents which require progression into S-phase of the cell-cycle for efficacy,
e.g.
radiation-induced death. Treatment by the subject method can provide
protection by
causing the hair follicle cells to become quiescent, e.g., by inhibiting the
cells from
entering S phase, and thereby preventing the follicle cells from undergoing
mitotic
catastrophe or programmed cell death. For instance, subject compounds can be
used for
patients undergoing chemo- or radiation-therapies which ordinarily result in
hair loss. By
inhibiting cell-cycle progression during such therapies, the subject treatment
can protect
hair follicle cells from death which might otherwise result from activation of
cell death
programs. After the therapy has concluded, the instant method can also be
removed with
concommitant relief of the inhibition of follicle cell proliferation.
The subject method can also be used in the treatment of folliculitis, such as
folliculitis decalvans, folliculitis ulerythematosa reticulata or keloid
folliculitis. For
example, a cosmetic prepration of a subject compound can be applied topically
in the
treatment of pseudofolliculitis, a chronic disorder occurring most often in
the
submandibular region of the neck and associated with shaving, the
characteristic lesions
of which are erythematous papules and pustules containing buried hairs.
In another aspect of the invention, the subject method can be used to induce
differentiation and/or inhibit proliferation of epithelially derived tissue.
Such forms of
these molecules can provide a basis for differentiation therapy for the
treatment of

CA 02386190 2002-04-11
WO 01/27135 PCT/US00/28479
-69-
hyperplastic and/or neoplastic conditions involving epithelial tissue. For
example, such
preparations can be used for the treatment of cutaneous diseases in which
there is
abnormal proliferation or growth of cells of the skin.
For instance, the pharmaceutical preparations of the invention are intended
for the
treatment of hyperplastic epidermal conditions, such as keratosis, as well as
for the
treatment of neoplastic epidermal conditions such as those characterized by a
high
proliferation rate for various skin cancers, as for example basal cell
carcinoma or
squamous cell carcinoma. The subject method can also be used in the treatment
of
autoimmune diseases affecting the skin, in particular, of dermatological
diseases
involving morbid proliferation and/or keratinization of the epidermis, as for
example,
caused by psoriasis or atopic dermatosis.
Many common diseases of the skin, such as psoriasis, squamous cell carcinoma,
keratoacanthoma and actinic keratosis are characterized by localized abnormal
proliferation and growth. For example, in psoriasis, which is characterized by
scaly, red,
elevated plaques on the skin, the keratinocytes are known to proliferate much
more
rapidly than normal and to differentiate less completely.
In one embodiment, the preparations of the present invention are suitable for
the
treatment of dermatological ailments linked to keratinization disorders
causing abnormal
proliferation of skin cells, which disorders may be marked by either
inflammatory or
non-inflammatory components. To illustrate, therapeutic preparations of a
subject
compound, e.g., which promotes quiescense or differentiation, can be used to
treat
varying forms of psoriasis, be they cutaneous, mucosal or ungual. Psoriasis,
as described
above, is typically characterized by epidermal keratinocytes which display
marked
proliferative activation and differentiation along a "regenerative" pathway.
Treatment
with an antiproliferative embodiment of the subject method can be used to
reverse the
pathological epidermal activiation and can provide a basis for sustained
remission of the
disease.
A variety of other keratotic lesions are also candidates for treatment with
the
subject method. Actinic keratoses, for example, are superficial inflammatory
premalignant tumors arising on sun-exposed and irradiated skin. The lesions
are
erythematous to brown with variable scaling. Current therapies include
excisional and

CA 02386190 2002-04-11
WO 01/27135 PCT/US00/28479
-70-
cryosurgery. These treatments are painful, however, and often produce
cosmetically
unacceptable scarring. Accordingly, treatment of keratosis, such as actinic
keratosis, can
include application, preferably topical, of a subject compound composition in
amounts
sufficient to inhibit hyperproliferation of epidermal/epidermoid cells of the
lesion.
Acne represents yet another dermatologic ailment which may be treated by the
subject method. Acne vulgaris, for instance, is a multifactorial disease most
commonly
occurring in teenagers and young adults, and is characterized by the
appearance of
inflammatory and noninflammatory lesions on the face and upper trunk. The
basic defect
which gives rise to acne vulgaris is hypercornification of the duct of a
hyperactive
sebaceous gland. Hypercornification blocks the normal mobility of skin and
follicle
microorganisms, and in so doing, stimulates the release of lipases by
Propinobacterium
acnes and Staphylococcus epidermidis bacteria and Pitrosporum ovale, a yeast.
Treatment with an antiproliferative subject compound, particularly topical
preparations,
may be useful for preventing the transitional features of the ducts, e.g.
hypercomification, which lead to lesion formation. The subject treatment may
further
include, for example, antibiotics, retinoids and antiandrogens.
The present invention also provides a method for treating various forms of
dennatitis. Dermatitis is a descriptive term referring to poorly demarcated
lesions which
are either pruritic, erythematous, scaly, blistered, weeping, fissured or
crusted. These
lesions arise from any of a wide variety of causes. The most common types of
dermatitis
are atopic, contact and diaper dermatitis. For instance, seborrheic dermatitis
is a chronic,
usually pruritic, dermatitis with erythema, dry, moist, or greasy scaling, and
yellow
crusted patches on various areas, especially the scalp, with exfoliation of an
excessive
amount of dry scales. The subject method can also be used in the treatment of
stasis
dermatitis, an often chronic, usually eczematous dermatitis. Actinic
dermatitis is
denmatitis that due to exposure to actinic radiation such as that from the
sun, ultraviolet
waves or x- or gamma-radiation. According to the present invention, the
subject method
can be used in the treatment and/or prevention of certain symptoms of
dermatitis caused
by unwanted proliferation of epithelial cells. Such therapies for these
various forms of
dermatitis can also include topical and systemic corticosteroids,
antipuritics, and
antibiotics.

CA 02386190 2002-04-11
WO 01/27135 PCT/US00/28479
-71-
Ailments which may be treated by the subject method are disorders specific to
non-humans, such as mange.
In still another embodiment, the subject method can be used in the treatment
of
human cancers, particularly basal cell carcinomas and other tumors of
epithelial tissues
such as the skin. For example, subject compounds can be employed, in the
subject
method, as part of a treatment for basal cell nevus syndrome (BCNS), and other
other
human carcinomas, adenocarcinomas, sarcomas and the like.
In a preferred embodiment, the subject method is used as part of a treatment
ot
prophylaxis regimen for treating (or preventing) basal cell carcinoma. The
deregulation
of the hedgehog signaling pathway may be a general feature of basal cell
carcinomas
caused by ptc mutations. Consistent overexpression of human ptc mRNA has been
described in tumors of familial and sporadic BCCs, determined by in situ
hybridization.
Mutations that inactivate ptc may be expected to result in overexpression of
mutant Ptc,
because ptc displays negative autoregulation. Prior research demonstrates that
overexpression of hedgehog proteins can also lead to tumorigenesis. That sonic
hedgehog (Shh) has a role in tumorigenesis in the mouse has been suggested by
research
in which transgenic mice overexpressing Shh in the skin developed features of
BCNS,
including multiple BCC-like epidermal proliferations over the entire skin
surface, after
only a few days of skin development. A mutation in the Shh human gene from a
BCC
was also described; it was suggested that Shh or other Hh genes in humans
could act as
dominant oncogenes in humans. Sporadic ptc mutations have also been observed
in
BCCs from otherwise nonmal individuals, some of which are UV-signature
mutations. In
one recent study of sporadic BCCs, five UV-signature type mutations, either CT
or
CCTT changes, were found out of fifteen tumors determined to contain ptc
mutations.
Another recent analysis of sporadic ptc mutations in BCCs and neuroectodermal
tumors
revealed one CT change in one of three ptc mutations found in the BCCs. See,
for
example, Goodrich et al. (1997) Science 277:1109-13; Xie et al. (1997) Cancer
Res
57:2369-72; Oro et al. (1997) Science 276:817-21; Xie et al. (1997) Genes
Chromosomes Cancer 18:305-9; Stone et al. (1996) Nature 384:129-34; and
Johnson et
al. (1996) Science 272:1668-71.

CA 02386190 2002-04-11
WO 01/27135 PCT/US00/28479
-72-
The subject method can also be used to treat patients with BCNS, e.g., to
prevent
BCC or other effects of the disease which may be the result of ptc loss-of-
function,
hedgehog gain-of-function, or smoothened gain-of-function. Basal cell nevus
syndrome
is a rare autosomal dominant disorder characterized by multiple BCCs that
appear at a
young age. BCNS patients are very susceptible to the development of these
tumors; in
the second decade of life, large numbers appear, mainly on sun-exposed areas
of the
skin. This disease also causes a number of developmental abnormalities,
including rib,
head and face alterations, and sometimes polydactyly, syndactyly, and spina
bifida. They
also develop a number of tumor types in addition to BCCs: fibromas of the
ovaries and
heart, cysts of the skin and jaws, and in the central nervous system,
medulloblastomas
and meningiomas. The subject method can be used to prevent or treat such tumor
types
in BCNS and non-BCNS patients. Studies of BCNS patients show that they have
both
genomic and sporadic mutations in the ptc gene, suggesting that these
mutations are the
ultimate cause of this disease.
In another aspect, the present invention provides pharmaceutical preparations
and
methods for controlling the formation of megakaryocyte-derived cells and/or
controlling
the functional performance of megakaryocyte-derived cells. For instance,
certain of the
compositions disclosed herein may be applied to the treatment or prevention of
a variety
hyperplastic or neoplastic conditions affecting platelets.
In another aspect, the present invention provides phannaceutical preparations
comprising the subject compounds. The compounds for use in the subject method
may
be conveniently formulated for administration with a biologically acceptable
and/or
sterile medium, such as water, buffered saline, polyol (for example, glycerol,
propylene
glycol, liquid polyethylene glycol and the like) or suitable mixtures thereof.
The
optimum concentration of the active ingredient(s) in the chosen medium can be
determined empirically, according to procedures well known to medicinal
chemists. As
used herein, "biologically acceptable medium" includes any and all solvents,
dispersion
media, and the like which may be appropriate for the desired route of
administration of
the pharmaceutical preparation. The use of such media for pharmaceutically
active
substances is known in the art. Except insofar as any conventional media or
agent is
incompatible with the activity of the subject compounds, its use in the
pharmaceutical

CA 02386190 2002-04-11
WO 01/27135 PCT/US00/28479
-73-
preparation of the invention is contemplated. Suitable vehicles and their
formulation
inclusive of other proteins are described, for example, in the book
Remington's
Pharmaceutical Sciences (Remington's Pharmaceutical Sciences. Mack Publishing
Company, Easton, Pa., USA 1985). These vehicles include injectable "deposit
formulations".
Pharmaceutical formulations of the present invention can also include
veterinary
compositions, e.g., pharmaceutical preparations of the subject compounds
suitable for
veterinary uses, e.g., for the treatment of live stock or domestic animals,
e.g., dogs.
Methods of introduction may also be provided by rechargeable or biodegradable
devices. Various slow release polymeric devices have been developed and tested
in vivo
in recent years for the controlled delivery of drugs, including proteinacious
biopharmaceuticals. A variety of biocompatible polymers (including hydrogels),
including both biodegradable and non-degradable polymers, can be used to form
an
implant for the sustained release of a subject compound at a particular target
site.
The preparations of the present invention may be given orally, parenterally,
topically, or rectally. They are of course given by forms suitable for each
administration
route. For example, they are administered in tablets or capsule form, by
injection,
inhalation, eye lotion, ointment, suppository, controlled release patch, etc.
administration
by injection, infusion or inhalation; topical by lotion or ointment; and
rectal by
suppositories. Oral and topical administrations are preferred.
The phrases "parenteral administration" and "administered parenterally" as
used
herein means modes of administration other than enteral and topical
administration,
usually by injection, and includes, without limitation, intravenous,
intramuscular,
intraarterial, intrathecal, intracapsular, intraorbital, intracardiac,
intradermal,
intraperitoneal, transtracheal, subcutaneous, subcuticular, intraarticulare,
subcapsular,
subarachnoid, intraspinal and intrasternal injection and infusion.
The phrases "systemic administration," "administered systemically,"
"peripheral
administration" and "administered peripherally" as used herein mean the
administration
of a compound, drug or other material other than directly into the central
nervous

CA 02386190 2002-04-11
WO 01/27135 PCT/US00/28479
-74-
system, such that it enters the patient's system and, thus, is subject to
metabolism and
other like processes, for example, subcutaneous administration.
These compounds may be administered to humans and other animals for therapy
by any suitable route of administration, including orally, nasally, as by, for
example, a
spray, rectally, intravaginally, parenterally, intracisternally and topically,
as by powders,
ointments or drops, including buccally and sublingually.
Regardless of the route of administration selected, the compounds of the
present
invention, which may be used in a suitable hydrated form, and/or the
pharmaceutical
compositions of the present invention, are formulated into pharmaceutically
acceptable
dosage forms such as described below or by other conventional methods known to
those
of skill in the art.
Actual dosage levels of the active ingredients in the pharmaceutical
compositions
of this invention may be varied so as to obtain an amount of the active
ingredient which
is effective to achieve the desired therapeutic response for a particular
patient,
composition, and mode of administration, without being toxic to the patient.
The selected dosage level will depend upon a variety of factors including the
activity of the particular compound of the present invention employed, or the
ester, salt
or amide thereof, the route of administration, the time of administration, the
rate of
excretion of the particular compound being employed, the duration of the
treatment,
other drugs, compounds and/or materials used in combination with the
particular
compound employed, the age, sex, weight, condition, general health and prior
medical
history of the patient being treated, and like factors well known in the
medical arts.
A physician or veterinarian having ordinary skill in the art can readily
detennine
and prescribe the effective amount of the pharmaceutical composition required.
For
example, the physician or veterinarian could start doses of the compounds of
the
invention employed in the pharmaceutical composition at levels lower than that
required
in order to achieve the desired therapeutic effect and gradually increase the
dosage until
the desired effect is achieved.
In general, a suitable daily dose of a compound of the invention will be that
amount of the compound which is the lowest dose effective to produce a
therapeutic

CA 02386190 2002-04-11
WO 01/27135 PCT/US00/28479
-75-
effect. Such an effective dose will generally depend upon the factors
described above.
Generally, intravenous, intracerebroventricular and subcutaneous doses of the
compounds of this invention for a patient will range from about 0.0001 to
about 100 mg
per kilogram of body weight per day.
If desired, the effective daily dose of the active compound may be
administered
as two, three, four, five, six or more sub-doses administered separately at
appropriate
intervals throughout the day, optionally, in unit dosage forms.
The term "treatment" is intended to encompass also prophylaxis, therapy and
cure.
The patient receiving this treatment is any animal in need, including
primates, in
particular humans, and other mammals such as equines, cattle, swine and sheep;
and
poultry and pets in general.
The compound of the invention can be administered as such or in admixtures
with pharmaceutically acceptable and/or sterile carriers and can also be
administered in
conjunction with other antimicrobial agents such as penicillins,
cephalosporins,
aminoglycosides and glycopeptides. Conjunctive therapy, thus includes
sequential,
simultaneous and separate administration of the active compound in a way that
the
therapeutical effects of the first administered one is not entirely
disappeared when the
subsequent is administered.
V. Pharmaceutical Compositions
While it is possible for a compound of the present invention to be
administered
alone, it is preferable to administer the compound as a pharmaceutical
formulation
(composition). The subject compounds according to the invention may be
formulated for
administration in any convenient way for use in human or veterinary medicine.
In certain
embodiments, the compound included in the pharmaceutical preparation may be
active
itself, or may be a prodrug, e.g., capable of being converted to an active
compound in a
physiological setting.
Thus, another aspect of the present invention provides pharmaceutically
acceptable compositions comprising a therapeutically effective amount of one
or more of

CA 02386190 2002-04-11
WO 01/27135 PCT/US00/28479
-76-
the compounds described above, formulated together with one or more
pharmaceutically
acceptable carriers (additives) and/or diluents. As described in detail below,
the
pharmaceutical compositions of the present invention may be specially
formulated for
administration in solid or liquid form, including those adapted for the
following: (1) oral
administration, for example, drenches (aqueous or non-aqueous solutions or
suspensions), tablets, boluses, powders, granules, pastes for application to
the tongue;
(2) parenteral administration, for example, by subcutaneous, intramuscular or
intravenous injection as, for example, a sterile solution or suspension; (3)
topical
application, for example, as a cream, ointment or spray applied to the skin;
or (4)
intravaginally or intrarectally, for example, as a pessary, cream or foam.
However, in
certain embodiments the subject compounds may be simply dissolved or suspended
in
sterile water. In certain embodiments, the pharmaceutical preparation is non-
pyrogenic,
i.e., does not elevate the body temperature of a patient.
The phrase "therapeutically effective amount" as used herein means that amount
of a compound, material, or composition comprising a compound of the present
invention which is effective for producing some desired therapeutic effect,
e.g., by
overcoming a ptc loss-of-function, hedgehog gain-of-function, or smoothened
gain-of-
function, in at least a sub-population of cells in an animal and thereby
blocking the
biological consequences of that pathway in the treated cells, at a reasonable
benefit/risk
ratio applicable to any medical treatment.
The phrase "pharmaceutically acceptable" is employed herein to refer to those
compounds, materials, compositions, and/or dosage forms which are, within the
scope of
sound medical judgment, suitable for use in contact with the tissues of human
beings and
animals without excessive toxicity, irritation, allergic response, or other
problem or
complication, commensurate with a reasonable benefit/risk ratio.
The phrase "pharmaceutically acceptable carrier" as used herein means a
pharmaceutically acceptable material, composition or vehicle, such as a liquid
or solid
filler, diluent, excipient, solvent or encapsulating material, involved in
carrying or
transporting the subject regulators from one organ, or portion of the body, to
another
organ, or portion of the body. Each carrier must be "acceptable" in the sense
of being
compatible with the other ingredients of the formulation and not injurious to
the patient.

CA 02386190 2002-04-11
WO 01/27135 PCT/USOO/28479
-77-
Some examples of materials which can serve as pharmaceutically acceptable
carriers
include: (1) sugars, such as lactose, glucose and sucrose; (2) starches, such
as corn starch
and potato starch; (3) cellulose, and its derivatives, such as sodium
carboxymethyl
cellulose, ethyl cellulose and cellulose acetate; (4) powdered tragacanth; (5)
malt; (6)
gelatin; (7) talc; (8) excipients, such as cocoa butter and suppository waxes;
(9) oils,
such as peanut oil, cottonseed oil, safflower oil, sesame oil, olive oil, corn
oil and
soybean oil; (10) glycols, such as propylene glycol; (11) polyols, such as
glycerin,
sorbitol, mannitol and polyethylene glycol; (12) esters, such as ethyl oleate
and ethyl
laurate; (13) agar; (14) buffering agents, such as magnesium hydroxide and
aluminum
hydroxide; (15) alginic acid; (16) pyrogen-free water; (17) isotonic saline;
(18) Ringer's
solution; (19) ethyl alcohol; (20) phosphate buffer solutions; and (21) other
non-toxic
compatible substances employed in pharmaceutical formulations.
As set out above, certain embodiments of the present compounds may contain a
basic functional group, such as amino or alkylamino, and are, thus, capable of
forming
pharmaceutically acceptable salts with pharmaceutically acceptable acids. The
term
"pharmaceutically acceptable salts" in this respect, refers to the relatively
non-toxic,
inorganic and organic acid addition salts of compounds of the present
invention. These
salts can be prepared in situ during the final isolation and purification of
the compounds
of the invention, or by separately reacting a purified compound of the
invention in its
free base form with a suitable organic or inorganic acid, and isolating the
salt thus
formed. Representative salts include the hydrobromide, hydrochloride, sulfate,
bisulfate,
phosphate, nitrate, acetate, valerate, oleate, palmitate, stearate, laurate,
benzoate, lactate,
phosphate, tosylate, citrate, maleate, fumarate, succinate, tartrate,
naphthylate, mesylate,
glucoheptonate, lactobionate, and laurylsulphonate salts and the like. (See,
for example,
Berge et al. (1977) "Pharmaceutical Salts", J. Pharm. Sci. 66:1-19)
The pharmaceutically acceptable salts of the subject compounds include the
conventional nontoxic salts or quaternary ammonium salts of the compounds,
e.g., from
non-toxic organic or inorganic acids. For example, such conventional nontoxic
salts
include those derived from inorganic acids such as hydrochloride, hydrobromic,
sulfuric,
sulfamic, phosphoric, nitric, and the like; and the salts prepared from
organic acids such
as acetic, propionic, succinic, glycolic, stearic, lactic, malic, tartaric,
citric, ascorbic,

CA 02386190 2002-04-11
WO 01/27135 PCT/US00/28479
-78-
palmitic, maleic, hydroxymaleic, phenylacetic, glutamic, benzoic, salicyclic,
sulfanilic,
2-acetoxybenzoic, fumaric, toluenesulfonic, methanesulfonic, ethane
disulfonic, oxalic,
isothionic, and the like.
In other cases, the compounds of the present invention may contain one or more
acidic functional groups and, thus, are capable of forming pharmaceutically
acceptable
salts with pharmaceutically acceptable bases. The term "pharmaceutically
acceptable
salts" in these instances refers to the relatively non-toxic, inorganic and
organic base
addition salts of compounds of the present invention. These salts can likewise
be
prepared in situ during the final isolation and purification of the compounds,
or by
separately reacting the purified compound in its free acid form with a
suitable base, such
as the hydroxide, carbonate or bicarbonate of a pharmaceutically acceptable
metal
cation, with ammonia, or with a pharmaceutically acceptable organic primary,
secondary
or tertiary amine. Representative alkali or alkaline earth salts include the
lithium,
sodium, potassium, calcium, magnesium, and aluminum salts and the like.
Representative organic amines useful for the formation of base addition salts
include
ethylamine, diethylamine, ethylenediamine, ethanolamine, diethanolamine,
piperazine
and the like. (See, for example, Berge et al., supra)
Wetting agents, emulsifiers and lubricants, such as sodium lauryl sulfate and
magnesium stearate, as well as coloring agents, release agents, coating
agents,
sweetening, flavoring and perfuming agents, preservatives and antioxidants can
also be
present in the compositions.
Examples of pharmaceutically acceptable antioxidants include: (1) water
soluble
antioxidants, such as ascorbic acid, cysteine hydrochloride, sodium bisulfate,
sodium
metabisulfite, sodium sulfite and the like; (2) oil-soluble antioxidants, such
as ascorbyl
palmitate, butylated hydroxyanisole (BHA), butylated hydroxytoluene (BHT),
lecithin,
propyl gallate, alpha-tocopherol, and the like; and (3) metal chelating
agents, such as
citric acid, ethylenediamine tetraacetic acid (EDTA), sorbitol, tartaric acid,
phosphoric
acid, and the like.
Formulations of the present invention include those suitable for oral, nasal,
topical (including buccal and sublingual), rectal, vaginal and/or parenteral
administration. The formulations may conveniently be presented in unit dosage
form and

CA 02386190 2002-04-11
WO 01/27135 PCT/US00/28479
-79-
may be prepared by any methods well known in the art of pharmacy. The amount
of
active ingredient which can be combined with a carrier material to produce a
single
dosage form will vary depending upon the host being treated, the particular
mode of
administration. The amount of active ingredient which can be combined with a
carrier
material to produce a single dosage form will generally be that amount of the
compound
which produces a therapeutic effect. Generally, out of one hundred per cent,
this amount
will range from about 1 per cent to about ninety-nine percent of active
ingredient,
preferably from about 5 per cent to about 70 per cent, most preferably from
about 10 per
cent to about 30 per cent.
Methods of preparing these formulations or compositions include the step of
bringing into association a compound of the present invention with the carrier
and,
optionally, one or more accessory ingredients. In general, the formulations
are prepared
by uniformly and intimately bringing into association a compound of the
present
invention with liquid carriers, or finely divided solid carriers, or both, and
then, if
necessary, shaping the product.
Formulations of the invention suitable for oral administration may be in the
form
of capsules, cachets, pills, tablets, lozenges (using a flavored basis,
usually sucrose and
acacia or tragacanth), powders, granules, or as a solution or a suspension in
an aqueous
or non-aqueous liquid, or as an oil-in-water or water-in-oil liquid emulsion,
or as an
elixir or syrup, or as pastilles (using an inert base, such as gelatin and
glycerin, or
sucrose and acacia) and/or as mouth washes and the like, each containing a
predetermined amount of a compound of the present invention as an active
ingredient. A
compound of the present invention may also be administered as a bolus,
electuary or
paste.
In solid dosage forms of the invention for oral administration (capsules,
tablets,
pills, dragees, powders, granules and the like), the active ingredient is
mixed with one or
more pharmaceutically acceptable carriers, such as sodium citrate or dicalcium
phosphate, and/or any of the following: (1) fillers or extenders, such as
starches, lactose,
sucrose, glucose, mannitol, and/or silicic acid; (2) binders, such as, for
example,
carboxymethylcellulose, alginates, gelatin, polyvinyl pyrrolidone, sucrose
and/or acacia;
(3) humectants, such as glycerol; (4) disintegrating agents, such as agar-
agar, calcium

CA 02386190 2002-04-11
WO 01/27135 PCT/US00/28479
-80-
carbonate, potato or tapioca starch, alginic acid, certain silicates, and
sodium carbonate;
(5) solution retarding agents, such as paraffin; (6) absorption accelerators,
such as
quatemary ammonium compounds; (7) wetting agents, such as, for example, cetyl
alcohol and glycerol monostearate; (8) absorbents, such as kaolin and
bentonite clay; (9)
lubricants, such a talc, calcium stearate, magnesium stearate, solid
polyethylene glycols,
sodium lauryl sulfate, and mixtures thereof; and (10) coloring agents. In the
case of
capsules, tablets and pills, the pharmaceutical compositions may also comprise
buffering
agents. Solid compositions of a similar type may also be employed as fillers
in soft and
hard-filled gelatin capsules using such excipients as lactose or milk sugars,
as well as
high molecular weight polyethylene glycols and the like.
A tablet may be made by compression or molding, optionally with one or more
accessory ingredients. Compressed tablets may be prepared using binder (for
example,
gelatin or hydroxypropylmethyl cellulose), lubricant, inert diluent,
preservative,
disintegrant (for example, sodium starch glycolate or cross-linked sodium
carboxymethyl cellulose), surface-active or dispersing agent. Molded tablets
may be
made by molding in a suitable machine a mixture of the powdered compound
moistened
with an inert liquid diluent.
The tablets, and other solid dosage forms of the pharmaceutical compositions
of
the present invention, such as dragees, capsules, pills and granules, may
optionally be
scored or prepared with coatings and shells, such as enteric coatings and
other coatings
well known in the pharmaceutical-formulating art. They may also be formulated
so as to
provide slow or controlled release of the active ingredient therein using, for
example,
hydroxypropylmethyl cellulose in varying proportions to provide the desired
release
profile, other polymer matrices, liposomes and/or microspheres. They may be
sterilized
by, for example, filtration through a bacteria-retaining filter, or by
incorporating
sterilizing agents in the form of sterile solid compositions which can be
dissolved in
sterile water, or some other sterile injectable medium immediately before use.
These
compositions may also optionally contain opacifying agents and may be of a
composition that they release the active ingredient(s) only, or
preferentially, in a certain
portion of the gastrointestinal tract, optionally, in a delayed manner.
Examples of
embedding compositions which can be used include polymeric substances and
waxes.

CA 02386190 2002-04-11
WO 01/27135 PCT/US00/28479
-81-
The active ingredient can also be in micro-encapsulated form, if appropriate,
with one or
more of the above-described excipients.
Liquid dosage forms for oral administration of the compounds of the invention
include pharmaceutically acceptable emulsions, microemulsions, solutions,
suspensions,
syrups and elixirs. In addition to the active ingredient, the liquid dosage
forms may
contain inert diluents commonly used in the art, such as, for example, water
or other
solvents, solubilizing agents and emulsifiers, such as ethyl alcohol,
isopropyl alcohol,
ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene
glycol, 1,3-
butylene glycol, oils (in particular, cottonseed, groundnut, corn, germ,
olive, castor and
sesame oils), glycerol, tetrahydrofuryl alcohol, polyethylene glycols and
fatty acid esters
of sorbitan, and mixtures thereof.
Besides inert diluents, the oral compositions can also include adjuvants such
as
wetting agents, emulsifying and suspending agents, sweetening, flavoring,
coloring,
perfuming and preservative agents.
Suspensions, in addition to the active compounds, may contain suspending
agents as, for example, ethoxylated isostearyl alcohols, polyoxyethylene
sorbitol and
sorbitan esters, microcrystalline cellulose, aluminum metahydroxide,
bentonite, agar-
agar and tragacanth, and mixtures thereof.
It is known that sterols, such as cholesterol, will form complexes with
cyclodextrins. Thus, in preferred embodiments, where the inhibitor is a
steroidal
alkaloid, it may be formulated with cyclodextrins, such as a-, (3- and y-
cyclodextrin,
dimethyl- (3 cyclodextrin and 2-hydroxypropyl-(3-cyclodextrin.
Formulations of the pharmaceutical compositions of the invention for rectal or
vaginal administration may be presented as a suppository, which may be
prepared by
mixing one or more compounds of the invention with one or more suitable
nonirritating
excipients or carriers comprising, for example, cocoa butter, polyethylene
glycol, a
suppository wax or a salicylate, and which is solid at room temperature, but
liquid at
body temperature and, therefore, will melt in the rectum or vaginal cavity and
release the
active compound.

CA 02386190 2002-04-11
WO 01/27135 PCT/US00/28479
-82-
Formulations of the present invention which are suitable for vaginal
administration also include pessaries, tampons, creams, gels, pastes, foams or
spray
formulations containing such carriers as are known in the art to be
appropriate.
Dosage forms for the topical or transdermal administration of a compound of
this
invention include powders, sprays, ointments, pastes, creams, lotions, gels,
solutions,
patches and inhalants. The active compound may be mixed under sterile
conditions with
a pharmaceutically acceptable carrier, and with any preservatives, buffers, or
propellants
which may be required.
The ointments, pastes, creams and gels may contain, in addition to an active
compound of this invention, excipients, such as animal and vegetable fats,
oils, waxes,
paraffins, starch, tragacanth, cellulose derivatives, polyethylene glycols,
silicones,
bentonites, silicic acid, talc and zinc oxide, or mixtures thereof.
Powders and sprays can contain, in addition to a compound of this invention,
excipients such as lactose, talc, silicic acid, aluminum hydroxide, calcium
silicates and
polyamide powder, or mixtures of these substances. Sprays can additionally
contain
customary propellants, such as chlorofluorohydrocarbons and volatile
unsubstituted
hydrocarbons, such as butane and propane.
Transdermal patches have the added advantage of providing controlled delivery
of a compound of the present invention to the body. Such dosage forms can be
made by
dissolving or dispersing the subject compounds in the proper medium.
Absorption
enhancers can also be used to increase the flux of the compound across the
skin. The rate
of such flux can be controlled by either providing a rate controlling membrane
or
dispersing the compound in a polymer matrix or gel.
Ophthalmic formulations, eye ointments, powders, solutions and the like, are
also
contemplated as being within the scope of this invention.
Pharmaceutical compositions of this invention suitable for parenteral
administration comprise one or more compounds of the invention in combination
with
one or more pharmaceutically acceptable sterile isotonic aqueous or nonaqueous
solutions, dispersions, suspensions or emulsions, or sterile powders which may
be
reconstituted into sterile injectable solutions or dispersions just prior to
use, which may

CA 02386190 2002-04-11
WO 01/27135 PCT/US00/28479
-83-
contain antioxidants, buffers, bacteriostats, solutes which render the
formulation isotonic
with the blood of the intended recipient or suspending or thickening agents.
Examples of suitable aqueous and nonaqueous carriers which may be employed
in the pharmaceutical compositions of the invention include water, ethanol,
polyols
(such as glycerol, propylene glycol, polyethylene glycol, and the like), and
suitable
mixtures thereof, vegetable oils, such as olive oil, and injectable organic
esters, such as
ethyl oleate. Proper fluidity can be maintained, for example, by the use of
coating
materials, such as lecithin, by the maintenance of the required particle size
in the case of
dispersions, and by the use of surfactants.
These compositions may also contain adjuvants such as preservatives, wetting
agents, emulsifying agents and dispersing agents. Prevention of the action of
microorganisms may be ensured by the inclusion of various antibacterial and
antifungal
agents, for example, paraben, chlorobutanol, phenol sorbic acid, and the like.
It may also
be desirable to include isotonic agents, such as sugars, sodium chloride, and
the like into
the compositions. In addition, prolonged absorption of the injectable
pharmaceutical
forrn may be brought about by the inclusion of agents which delay absorption
such as
aluminum monostearate and gelatin.
In some cases, in order to prolong the effect of a drug, it is desirable to
slow the
absorption of the drug from subcutaneous or intramuscular injection. This may
be
accomplished by the use of a liquid suspension of crystalline or amorphous
material
having poor water solubility. The rate of absorption of the drug then depends
upon its
rate of dissolution which, in turn, may depend upon crystal size and
crystalline form.
Alternatively, delayed absorption of a parenterally administered drug form is
accomplished by dissolving or suspending the drug in an oil vehicle.
Injectable depot forms are made by forming microencapsule matrices of the
subject compounds in biodegradable polymers such as polylactide-polyglycolide.
Depending on the ratio of drug to polymer, and the nature of the particular
polymer
employed, the rate of drug release can be controlled. Examples of other
biodegradable
polymers include poly(orthoesters) and poly(anhydrides). Depot injectable
formulations
are also prepared by entrapping the drug in liposomes or microemulsions which
are
compatible with body tissue.

CA 02386190 2002-04-11
WO 01/27135 PCT/US00/28479
-84-
When the compounds of the present invention are administered as
pharmaceuticals, to humans and animals, they can be given per se or as a
pharmaceutical
composition containing, for example, 0.1 to 99.5% (more preferably, 0.5 to
90%) of
active ingredient in combination with a pharmaceutically acceptable carrier.
The addition of the active compound of the invention to animal feed is
preferably
accomplished by preparing an appropriate feed premix containing the active
compound
in an effective amount and incorporating the premix into the complete ration.
Alternatively, an intermediate concentrate or feed supplement containing the
active ingredient can be blended into the feed. The way in which such feed
premixes and
complete rations can be prepared and administered are described in reference
books
(such as "Applied Animal Nutrition", W.H. Freedman and CO., San Francisco,
U.S.A.,
1969 or "Livestock Feeds and Feeding" 0 and B books, Corvallis, Ore., U.S.A.,
1977).
VI. Synthetic Schemes and Identi tcation ofActive ReQulators
The subject compounds, and derivatives thereof, can be prepared readily by
employing known synthetic methodology. As is well known in the art, these
coupling
reactions are carried out under relatively mild conditions and tolerate a wide
range of
"spectator" functionality. Additional compounds may be synthesized and tested
in a
combinatorial fashion, to facilitate the identification of additional
compounds which
may be employed in the subject method.
a. Combinatorial Libraries
The compounds of the present invention, particularly libraries of variants
having
various representative classes of substituents, are amenable to combinatorial
chemistry
and other parallel synthesis schemes (see, for example, PCT WO 94/08051). The
result
is that large libraries of related compounds, e.g. a variegated library of
compounds
represented above, can be screened rapidly in high throughput assays in order
to identify
potential hedgehog regulator lead compounds, as well as to refine the
specificity,
toxicity, and/or cytotoxic-kinetic profile of a lead compound. For instance,
ptc,
hedgehog, or smoothened bioactivity assays, such as may be developed using
cells with

CA 02386190 2002-04-11
WO 01/27135 PCT/US00/28479
-85-
either a ptc loss-of-function, hedgehog gain-of-function, or smoothened gain-
of-function,
can be used to screen a library of the subject compounds for those having
agonist
activity toward ptc or antagonist activity towards hedgehog or smoothened.
Alternatively, bioactivity assays using cells with either a ptc gain-of-
function, hedgehog
loss-of-function, or smoothened loss-of-function, can be used to screen a
library of the
subject compounds for those having antagonist activity toward ptc or agonist
activity
towards hedgehog or smoothened.
Simply for illustration, a combinatorial library for the purposes of the
present
invention is a mixture of chemically related compounds which may be screened
together
for a desired property. The preparation of many related compounds in a single
reaction
greatly reduces and simplifies the number of screening processes which need to
be
carried out. Screening for the appropriate physical properties can be done by
conventional methods.
Diversity in the library can be created at a variety of different levels. For
instance, the substrate aryl groups used in the combinatorial reactions can be
diverse in
terms of the core aryl moiety, e.g., a variegation in terms of the ring
structure, and/or can
be varied with respect to the other substituents.
A variety of techniques are available in the art for generating combinatorial
libraries of small organic molecules such as the subject compounds. See, for
example,
Blondelle et al. (1995) Trends Anal. Chem. 14:83; the Affymax U.S. Patents
5,359,115
and 5,362,899: the Ellman U.S. Patent 5,288,514: the Still et al. PCT
publication WO
94/08051; the ArQule U.S. Patents 5,736,412 and 5,712,171; Chen et al. (1994)
JACS
116:2661: Kerr et al. (1993) JACS 115:252; PCT publications W092/10092,
W093/09668 and W091/07087; and the Lerner et al. PCT publication W093/20242).
Accordingly, a variety of libraries on the order of about 100 to 1,000,000 or
more
diversomers of the subject compounds can be synthesized and screened for
particular
activity or property.
In an exemplary embodiment, a library of candidate compound
diversomers can be synthesized utilizing a scheme adapted to the techniques
described in

CA 02386190 2002-04-11
WO 01/27135 PCT/US00/28479
-86-
the Still et al. PCT publication WO 94/08051, e.g., being linked to a polymer
bead by a
hydrolyzable or photolyzable group, optionally located at one of the positions
of the
candidate regulators or a substituent of a synthetic intermediate. According
to the Still et
al. technique, the library is synthesized on a set of beads, each bead
including a set of
tags identifying the particular diversomer on that bead. The bead library can
then be
"plated" with, for example, ptc loss-of-function, hedgehog gain-of-function,
or
smoothened gain-of-function cells for which a smoothened antagonist is sought.
The
diversomers can be released from the bead, e.g. by hydrolysis.
Many variations on the above and related pathways permit the synthesis of
widely diverse libraries of compounds which may be tested as regulators of
hedgehog
function.
b. Screening Assays
There are a variety of assays available for determining the ability of a
compound
such as a hedgehog regulator to regulate ptc, smoothened, or hedgehog
function, many
of which can be disposed in high-throughput formats. In many drug screening
programs
which test libraries of compounds and natural extracts, high throughput assays
are
desirable in order to maximize the number of compounds surveyed in a given
period of
time. Thus, libraries of synthetic and natural products can be sampled for
other
compounds which are hedgehog regulators.
In addition to cell-free assays, test compounds can also be tested in cell-
based
assays. In one embodiment, cells which have a ptc loss-of-function, hedgehog
gain-of-
function, or smoothened gain-of-function phenotype can be contacted with a
test agent
of interest, with the assay scoring for, e.g., inhibition of proliferation of
the cell in the
presence of the test agent.
A number of gene products have been implicated in patched-mediated signal
transduction, including patched, transcription factors of the cubitus
interruptus (ci)
family, the serine/threonine kinase fused (fu) and the gene products of costal-
2,
smoothened and suppressor offused.

CA 02386190 2002-04-11
WO 01/27135 PCT/US00/28479
-87-
The induction of cells by hedgehog proteins sets in motion a cascade involving
the activation and inhibition of downstream effectors, the ultimate
consequence of which
is, in some instances, a detectable change in the transcription or translation
of a gene.
Potential transcriptional targets of hedgehog-mediated signaling are the
patched gene
(Hidalgo and Ingham, 1990 Development 110, 291-301; Marigo et al., 1996 ) and
the
vertebrate homologs of the drosophila cubitus interruptus gene, the GLI genes
(Hui et al.
(1994) Dev Biol 162:402-413). Patched gene expression has been shown to be
induced
in cells of the limb bud and the neural plate that are responsive to Shh.
(Marigo et al.
(1996) PNAS 93:9346-51; Marigo et al. (1996) Development 122:1225-1233). The
Gli
genes encode putative transcription factors having zinc finger DNA binding
domains
(Orenic et al. (1990) Genes & Dev 4:1053-1067; Kinzler et al. (1990) Mol Cell
Biol
10:634-642). Transcription of the Gli gene has been reported to be upregulated
in
response to hedgehog in limb buds, while transcription of the Gli3 gene is
downregulated in response to hedgehog induction (Marigo et al. (1996)
Development
122:1225-1233). By selecting transcriptional regulatory sequences from such
target
genes, e.g., from patched or Gli genes, that are responsible for the up- or
down-
regulation of these genes in response to hedgehog signalling, and operatively
linking
such promoters to a reporter gene, one can derive a transcription based assay
which is
sensitive to the ability of a specific test compound to modify hedgehog-
mediated
signalling pathways. Expression of the reporter gene, thus, provides a
valuable screening
tool for the development of compounds that act as regulators of hedgehog. '
Reporter gene based assays of this invention measure the end stage of the
above
described cascade of events, e.g., transcriptional modulation. Accordingly, in
practicing
one embodiment of the assay, a reporter gene construct is inserted into the
reagent cell in
order to generate a detection signal dependent on ptc loss-of-function,
hedgehog gain-of-
function, smoothened gain-of-function, or stimulation by Shh itself. The
amount of
transcription from the reporter gene may be measured using any method known to
those
of skill in the art to be suitable. For example, mRNA expression from the
reporter gene
may be detected using RNAse protection or RNA-based PCR, or the protein
product of
the reporter gene may be identified by a characteristic stain or an intrinsic
biological
activity. The amount of expression from the reporter gene is then compared to
the
amount of expression in either the same cell in the absence of the test
compound or it

CA 02386190 2002-04-11
WO 01/27135 PCT/US00/28479
-88-
may be compared with the amount of transcription in a substantially identical
cell that
lacks the target receptor protein. Any statistically or otherwise significant
decrease in the
amount of transcription indicates that the test compound has in some manner
agonized
the normal ptc signal (or antagonized the gain-of-function hedgehog or
smoothened
signal), e.g., the test compound is a potential smoothened antagonist.
Exemplification
The invention now being generally described, it will be more readily
understood
by reference to the following examples which are included merely for purposes
of
illustration of certain aspects and embodiments of the present invention, and
are not
intended to limit the invention.
Example 1: Steroidal Compounds
Hedgehog signaling pathways are required for normal embryonic development
yet also are associated with carcinogenesis (L. V. Goodrich and M. P. Scott,
Neuron 21,
1243 (1998)). Loss of Sonic hedgehog signaling (Shh), for example, can result
in
cyclopia and other developmental defects of the face, forebrain, and other
organs and
structures (C. Chiang et al., Nature 383, 407 (1996)), whereas inappropriate
activation of
the pathway is associated with basal cell carcinoma, medulloblastoma, and
other
neoplastic disorders (H. Hahn et al., Cell 85, 841 (1996); R. L. Johnson et
al., Science
272, 1668 (1996); M. Gailani et al., Nat Genet 14, 78 (1996); T. Pietsch et
al., Cancer
Res 57, 2085 (1997); J. Reifenberger et al., Cancer Res. 58, 1798 (1998); C.
Raffel et al.,
Cancer Res 57, 842 (1997); J. Xie et al., Nature 391, 90 (1998)).
Pharmacological
manipulation of this pathway thus might help elucidate the mechanisms of
signal
transduction and also provide a practical means to prevent or remedy somatic
and
congenital abnormalities. Cyclopamine, a plant steroidal alkaloid, has long
been known
to induce cyclopia and other manifestations of severe HPE in vertebrate
embryos (R. F.
Keeler and W. Binns, Teratology 1, 5 (1968)) and more recently was shown to
act by
inhibiting the cellular response to the Shh signal (M. K. Cooper, J. A.
Porter, K. E.

CA 02386190 2002-04-11
WO 01/27135 PCT/US00/28479
-89-
Young, P. A. Beachy, Science 280, 1603 (1998); J. P. Incardona, W. Gaffield,
R. P.
Kapur, H. Roelink, Development 125, 3553 (1998)). To evaluate the therapeutic
potential of cyclopamine, we investigated the mechanism by which it acts.
Cellular responses to the Hh signal are controlled by two multi-pass
transmembrane proteins, Smo and Ptc 1, predicted to have seven and twelve
transmembrane spans, respectively. Smo is related to the Frizzled family of
Wnt
receptors and more distantly to the secretin family of G protein-coupled
receptors (M. R.
Barnes, D. M. Duckworth, L. J. Beeley, Trends Pharmacol Sci 19, 399 (1998)).
Genetic
and biochemical evidence suggests that Ptc suppresses the activity of Smo, and
that Hh
binding to Ptc relieves this suppression, allowing activation of downstream
targets
through the Ci/GLI family of transcriptional effectors ( P. Aza-Blanc, F.-A.
Remirez-
Weber, M.-P. Laget, C. Schwartz, T. B. Komberg, Cell 89, 1043 (1997); N.
Methot and
K. Basler, Cell 96, 819 (1999); C. H. Chen et al., Cell 98, 305 (1999); B.
Wang, J.
Fallon, P. Beachy, Cell 100, 423 (2000)). To establish a cultured cell based
assay that is
sensitive to cyclopamine (a previously established Hedgehog signaling assay in
Drosophila cl-8 cells (Chen et al., 1999) is resistant to cyclopamine (not
shown)), we
screened several vertebrate cell lines for a transcriptional response to fully
modified
ShhNp (Fig. 2A) using a luciferase reporter driven by a promoter comprising
eight
synthetic Gli binding sites fused to the lens crystallin minimal promoter (H.
Sasaki, C.-
C. Hui, M. Nakafuku, H. Kondoh, Development 124, 1313 (1997)). Among several
fibroblast cell lines that respond to ShhNp, NIH-3T3 mouse embryonic
fibroblasts,
which respond with a 20-150 fold induction of luciferase activity (Fig. 2B),
were
selected for further studies. Importantly, when the cells were treated with
cyclopamine,
this induction by ShhNp was completely abolished (Fig. 2C).
Similar to the Hh signaling assay in Drosophila cl-8 cells, the response to
induction by ShhNp required functional Gli binding sites in the reporter (not
shown), and
the response was augmented by overexpression of Smo and suppressed by
overexpression of Ptc or of activated PKA (Table 1). Pharmacological
activation of
endogenous PKA by forskolin also prevented induction of reporter expression
(Table 1).
Using this assay, we confirmed the results of Xie et al. that the tumor
mutation W539L
(SmoAl) constitutively activates Smo, and found that another mutation from
tumor
tissue, S537N (SmoA2) also activates Smo. Expression of either of the
activating

CA 02386190 2002-04-11
WO 01/27135 PCT/US00/28479
-90-
mutants induced reporter expression to a level comparable to that observed in
ShhNp-
treated cells. To further examine the validity of this signaling assay we
transfected cells
with constructs encoding known pathway components or treated cells with known
inhibitors of the pathway and analyzed the effects of these treatments, alone
and in
combination, on reporter activation (Table 1). The major findings of
Drosophila and
mouse genetic analyses were confirmed, indicating that NIH-3T3 cells provide a
faithful
and physiologically meaningful model for analysis of the Shh signaling
pathway.
Table 1
Luciferase activityfrom a Gli-dependent reporter as induced by combinations of
Shh pathway inducing
and suppressing treatments
Inducer Suppressor
None Ptc cyclopamine forskolin PKA Gli3-N
None - - - - - -
ShhNp +++ + - - - -
Smo + - - - - ND
activated Smo +++ +* +* - - -
G1i2 ++ ++ ++ ++ + -
* Higher than normal dose required to completely suppress
G1i3-N = Gli3 truncated at residue 700, generating a repressor form
Expression constructs used: pRK5 for full length mouse Ptcl, C-terminally
truncated Ptcl-CTD, Gli3(1-
700) repressor and active PKA cDNAs, andpGE (transient transfections) or
pcDNA3.1 +hygro (stable
lines; Invitrogen) for the various Smo cDNAs. The CMV promoter driven
mammalian expression vector
pGE was derived from pEGFP-C1 (Clontech) by removing the sequences encoding
the EGFP. Renilla
luciferase (pRL-TK) was fused to the C-terminus of the Smo open reading frame.
The resulting fusion
protein constructs had comparable activity to corresponding untagged
constructs in the NIH-3 T3 assay
We then made several clonal NIH-3T3 cell lines that contain stably integrated
reporter. In the best-responding cell lines, we observed 20-60 fold induction
of luciferase
activity by ShhNp. Using these cell lines, we found that a full response to
ShhNp
required that cells have reached saturation density, with a reduction in
response observed
for less dense cultures (Fig. 2D). The requirement for saturation density also
applied to
Shh response in NIH-3T3 cells transiently transfected with luciferase reporter
(not

CA 02386190 2002-04-11
WO 01/27135 PCT/US00/28479
-91-
shown), and to reporter activation by expression of activated Smo (Fig. 2D),
but not to
induction by Gli 1 overexpression (not shown).
The steroidal nature of these plant teratogens and their ability to disrupt
cholesterol synthesis and/or transport (P. A. Beachy et al., Cold Spring Harb
Symp
Quant Biol 62, 191 (1997); Y. Lange, J. Ye, M. Rigney, T. L. Steck, JLipid Res
40,
2264 (1999)) suggested the possibility that they may affect the action of
Ptc1, which
contains an apparent sterol sensing domain (S. K. Loftus et al., Science 277,
232 (1997)).
Having established the characteristics of Shh reponse and cyclopamine
inhibition in
mouse embryonic fibroblasts, we assayed fibroblasts derived from Ptcl -/-
mouse
embryos for sensitivity to cyclopamine. Mice lacking function of Ptc1 display
widespread activation of targets of Shh signaling, including of the Ptcl gene
itself (L. V.
Goodrich, L. Milenkovic, K. M. Higgins, M. P. Scott, Science 277, 1109
(1997)). As (3-
galactosidase is expressed under the control of the Ptcl promoter in these
cells, 0-
galactosidase expression can be used to assay the state of Shh pathway
activity (Fig.
3A). Surprisingly, addition of cyclopamine to Ptc1-l- cells significantly
suppressed R-
galactosidase expression (Fig. 3A) and similarly suppressed activity of the
Gli-Luc
reporter (not shown), indicating that cyclopamine is able to suppress Shh
pathway
activity in the absence of Ptc 1 function. In contrast, cyclopamine failed to
prevent
pathway activation induced by G1i2 overexpression (Table 1). These results
suggest that
the target of cyclopamine action is not Ptcl (Ptc2 is not a likely target of
cyclopamine in
Ptcl-/- cells, as expression of the Ptc2 protein suppresses pathway activation
and the
pathway is maximally activated in Ptcl -/- fibroblasts (data not shown)), but
likely
another pathway component that functions somewhere between Ptc 1 and the Gli
proteins.
To further investigate the site of cyclopamine action we transfected NIH-3T3
cells with Smo cDNA (a mouse Smo cDNA probe was generated using RT-PCR with
degenerate oligonucleotide primers based on rat and human Smo sequences; this
probe
was subsequently used to isolate a cDNA clone containing the complete coding
sequence of mouse Smo), and found that overexpression of Smo in the absence of
Shh
induces reporter expression -10-fold. As this activation of the pathway occurs
in the
absence of Shh and can be suppressed by 5 M cyclopamine (Fig. 3B), we infer
that the
mechanism of cyclopamine action is not direct interference with Shh binding
(i.e., as a

CA 02386190 2002-04-11
WO 01/27135 PCT/US00/28479
-92-
neutral antagonist of Shh). Interestingly, cyclopamine at this concentration
showed little
effect on reporter expression induced by the tumor-derived activated Smo
mutants (Fig.
21 B), suggesting the possibility that cyclopamine acts directly or indirectly
upon Smo
and that activating mutations render Smo proteins resistant. Cyclopamine
resistance of
SmoAl also was observed at sub-maximal levels of pathway activation associated
with
reduced SmoAl expression (Fig. 3B). In addition, we tested whether Shh
signaling
through activated Smo is affected by cyclopamine. Although activated Smo
proteins
previously have been reported to resist suppression by Ptc1 (M. Murone, A.
Rosenthal,
F. J. de Sauvage, Current Biol. 9, 76 (1999)) we found that this resistance is
partial, as
transfection of a 9 to 1 ratio of a Ptc 1 construct (not shown) or of Ptc 1-
CTD, a C-
terminally deleted construct (Ptc 1-CTD previously was shown to be expresed at
higher
levels than Ptcl; N. Fuse et al., Proc Natl Acad Sci USA 96, 10992 (1999)),
can
completely inhibit the activating effects of SmoAl or SmoA2 (Fig. 3C). In
cells thus
transfected, the Gli-responsive reporter can be induced upon treatment with
ShhNp;
induction under these circumstances is resistant to 5 M cyclopamine (Fig.
3C), which
normally would abolish Shh signaling. These results indicate that activated
Smo
molecules in the presence of sufficiently high levels of Ptcl can contribute
to an
essentially normal, albeit cyclopamine resistant, response to the Shh signal.
The
requirement for higher levels of Ptc 1 is not due simply to a higher level of
the Smo
protein variant, as we found that the levels of wild type and activated Smo
proteins
produced in transfected cells were similar, despite dramatically elevated
levels of
reporter activity associated with activated Smo (Fig. 3D).
Although increasing levels of cyclopamine produce some inhibition of activated
Smo (although little if any inhibitory effect on activated Smo is observed at
3 M
cyclopamine (not shown), a concentration sufficient to completely inhibit
normal Shh
signaling, some inhibitory effect of 5 M cyclopamine is demonstrated in Fig.
4B),
complete inhibition was precluded by the toxic effects of cyclopamine that
emerge in the
10-40 M range (data not shown). However, a chemically synthesized cyclopamine
derivative, 3-keto, N-aminoethyl aminocaproyl dihydrocinnamoyl cyclopamine
(KAAD
cyclopamine, compound 33), displayed 10-20 fold greater potency in suppression
of
ShhNp-induced pathway activity while maintaining similar or lower toxicity
(Fig. 4A).
This compound suppressed SmoA 1-induced reporter activity at a concentration

CA 02386190 2002-04-11
WO 01/27135 PCT/US00/28479
- 93 -
approximately 10-fold higher than that required for suppression of ShhNp
signaling (Fig.
4A) and also displayed higher potency than cyclopamine in P2PTC_/_ cells (Fig.
4B). This
more potent cyclopamine derivative thus suppresses activated Smo as
effectively as high
levels of Ptcl.
The simplest explanation of cyclopamine resistance as conferred by activated
Smo proteins is that cyclopamine affects Smo activity and that activating
mutations
render Smo proteins resistant. An alternative interpretation would be that
activated Smo
proteins produce a high abundance of a downstream component and that a high
cyclopamine level is required to suppress the increased concentration of this
downstream
component. This alternative model, however, can not account for the sustained
cyclopamine resistance of activated Smo proteins observed at intermediate or
low levels
of pathway activation (Fig. 3B) (production by activated Smo of high levels of
a
downstream component that is the cyclopamine target can not explain the
sustained
cyclopamine resistance observed at intermediate levels of pathway activation
(Fig. 3B),
as the hypothetical cyclopamine target in this circumstance would be present
at the same
moderate levels as those produced by ShhNp signaling via unaltered Smo. We
also find
that high levels of Smo in maximally-stimulated cells do not confer
cyclopamine
resistance (not shown), again inconsistent with the notion that extensive
production or
activation by Smo of a downstream component can confer cyclopamine
resistance). As
activated Smo is not expressed at higher levels than unaltered Smo (Fig. 3D),
it would
appear that activating mutations may confer a higher intrinsic ability to
activate the
pathway. This suggests that, like other seven transmembrane receptors (R. A.
Bond et
al., Nature 374, 272 (1995); H. R. Bourne, Curr Opin Cell Bio19, 134 (1997)),
Smo may
exist in a balance between active and inactive forms. Cyclopamine and Ptc
activities
might shift this balance toward the inactive state and tumor-associated
mutations toward
the active state, thus accounting for the higher levels of Ptc and cyclopamine
activity
required to suppress activated Smo proteins.
Cyclopamine appears to impact the Shh pathway at the level of Smo activity
(see
above), but this action need not be direct and could operate through an effect
on
molecules involved in intracellular transport, on molecules that affect
posttranslational
modification of Smo, or on other molecules that impact Smo activity. Such
indirect
action of cyclopamine would not necessarily be inconsistent with a
conformational

CA 02386190 2002-04-11
WO 01/27135 PCT/US00/28479
-94-
transition between active and inactive Smo, as conformational state could be
coupled by
a variety of mechanisms to subcellular localization or state of covalent
modification.
Whatever the mechanism, such inhibitors may have utility in treament of
disorders
caused by inappropriate Shh pathway activation. Patients with Basal Cell Nevus
Syndrome (also termed Gorlin's syndrome), an autosomal dominant disorder
associated
with heterozygous loss-of-function mutations in the human Ptcl gene, display
increased
incidence of numerous tumors, most notably basal cell carcinoma (BCC),
medulloblastoma, rhabdomyosarcoma and fibrosarcoma. Loss-of-function mutations
in
Ptcl or activating mutations in Smo in addition are found in -40% of sporadic
BCC and
-25% of primitive neuroectodermal tumors. The ability of cyclopamine and its
derivatives to block pathway activation by both of these types of mutations
suggests that
these plant-derived compounds or others that influence the activity of Smo may
be
valuable as therapeutic agents.
Example 2: Steroid Derivatives
New derivatives synthesized
Cyclopamine and jervine (structures 1 and 2 of Figure 5, respectively) are
closely
related plant-derived steroidal alkaloids known to specifically inhibit the
Sonic
hedgehog signaling pathway (Cooper et al., Science 280, p. 1603-1607, 1998).
We have
synthesized chemically 23 new derivatives of these two compounds, by various
modifications of its secondary amine, the C-3 oxygen, and/or the C5-C6 olefin.
Some of
these compounds can be readily synthesized in labelled form, thus making them
useful
for binding studies. Some of these compounds also contain functional groups
useful for
photo-activatable cross-linking and consequent radiolabelling or attachment of
a biotin
moiety to target proteins. One of the compounds is fluorescently labelled, and
may be
useful for direct observation of the cellular target of cyclopamine action.
The potency of
the various derivatives is set forth in Table II.
Table II
Compound ICSa Compound IC50
1 >l00 nM 33 >lO nM

CA 02386190 2002-04-11
WO 01/27135 PCT/US00/28479
-95-
2 >l00 nM 34 >10 nM
3 >1 M 39 >100 nM
12 >10 M 40 >100 nM
18 >100nM 41 >100nM
19 >10nM 42 >100nM
20 > 100 nM 43 > 100 nM
21 >10 nM 50 >1 M
22 >l M 51 >1 M
26 >100 nM 57 >10 M
29 >100 nM 58 >10 M
30 >100 nM
Realization of improved potencies through SAR studies
Using a clonal cell line derived from parental NIH-3T3 fibroblast cells that
contains a stably integrated Shh-responsive luciferase reporter, we have
determined the
concentration of each compound required to achieve 50% inhibition of Sonic
hedgehog
induction (IC50). Through SAR studies we have found that adducts to the 3,B-
hydroxyl
dramatically reduce activity, whereas oxidation of the 3,f3-hydroxyl to a keto
group
increases potency. We have also found that addition of bulky groups to the
secondary
amine reduces potency, but that longer aliphatic linkers not only permit
addition of such
bulky groups but also enhance potency. The most active compound thus far
identified
(structure 34 in Figure 5; IC50 = 30 nM) displays a potency ten-fold greater
than that of
cyclopamine (structure 1; IC50 = 300 nM). It should be straighforward to
achieve even
greater potencies by systematically testing various adducts to the secondary
amine and
combining these in combination with a 3-keto functionality. The more potent
derivatives
we have already synthesized display the desirable property of achieving
inhibition of
Shh signaling with much reduced toxicity as compared to the parent compounds.
Broad utility of compounds
We have determined that these compounds are capable of blocking pathway
activity in cells with elevated levels of pathway activity due to lack of
function of the

CA 02386190 2002-04-11
WO 01/27135 PCT/US00/28479
-96-
Patched 1(Ptc 1) protein, or to constitutively activated function of the Smo
protein. The
ability of these compounds to block pathway activity in cells with both of
these types of
defects suggests that they may be broadly useful in the treatment of certain
sporadic
tumors or in prophylactic treatment of patients with an inherited disposition
to high
frequency formation of these tumors. Such tumors include but are not limited
to basal
cell carcinoma, medulloblastoma, fibrosarcoma, and rhabdomyosarcoma.
Additional
applications for these compounds include but are not limited to induction of
pancreatic
tissue, elimination of excessive hair growth, and treatment of other skin
disorders.
Response to the Hh signal is controlled by two transmembrane proteins, Patched
(Ptc) and Smoothened (Smo). Ptc is a twelve-span transmembrane protein that
binds
directly to Hh. Smo contains seven transmembrane spans, is related to the
Frizzled
family of Wnt receptors, and is related more distantly to members of the G-
protein
coupled receptor family. Genetic evidence indicates that Ptc suppresses the
activity of
Smo; Hh relieves this suppression and allows activation of downstream targets,
including the Ci/GLI family of transcriptional effectors.
The Ptcl protein is involved in suppressing pathway activity, and cells
lacking it
display constitutively high levels of pathway activation. A lack of Ptcl
function is
causally associated with a high percentage of sporadic basal cell carcinoma,
medulloblastoma, fibrosarcoma, rhabdomyosarcoma, as well as other tumors. In
addition
familial heterozygosity at the Ptcl locus is associated with a predisposition
to high
frequency formation of these tumors. We have shown that cyclopamine, jervine,
and
related compounds are capable of fully suppressing pathway activity in cells
lacking Ptc
function.
The Smo protein is required for pathway activation, and certain mutations of
the
Smo locus result in constitutive pathway activity, even in the absence of Shh
stimulation. We have found that cells expressing such activated Smo proteins
are
somewhat resistant to these compounds, but can still be suppressed at levels
between one
and two orders of magnitude higher than those required for suppression of
normal cells
stimulated with Shh protein. Thus, tumors associated with sporadic activating
mutations
at the Smo locus may also be responsive to treatment with these compounds.

CA 02386190 2002-04-11
WO 01/27135 PCT/US00/28479
-97-
Summary of structure-activity-relationship (SAR) data. Modifications to
cyclopamine (1) and jervine (2) are most efficiently accomplished at
heteroatom
positions, namely the secondary amine and the secondary alcohol. Our initial
investigations of alkaloid derivatives therefore focused upon the chemical
synthesis of
such heteroatom modifications and the biological evaluation of these novel
compounds.
Using standard synthetic procedures and a cell-based Shh signaling assay, it
was
determined that the conjugation of chemical groups to the steroid alcohol
through a
carbamate linkage effectively diminishes Shh inhibitory activity; C3-OH
modified
compounds 8 and 12 exhibit IC50s in the cell-based assay that are nearly 100
times
higher than those of cyclopamine. These carbamate-containing compounds are
also 10
times weaker than cycloposine (3), which has a more labile modification of the
C3-OH
group, suggesting that the observed inhibitory activity of cycloposine is
actually due to
partial hydrolysis of the glycosidic linkage.
In contrast, certain modifications of the secondary amine that preserve the
basicity of this moiety are accommodated by the cyclopamine target. All N-
alkyl
derivatives in this study were synthetically obtained through the diamine
intermediate
17, which allows for the efficient preparation of numerous cyclopamine
analogs. Even
medium-sized structural elements such as those found in compounds 29, 33, 39,
40, and
43 do not significantly diminish the abilities of these alkaloids to block Shh
signaling,
and most additions even appear to accentuate inhibitory activity (29, 33, 39,
and 43).
These observations are somewhat different from those reported by Keeler and co-
workers, in which small N-alkyl derivatives of cyclopamine demonstrated
diminished
teratogenic potential in live animals. The reasons for these differences are
unclear,
although they could reflect metabolic and/or pharmacologic influences in the
animal-
based studies. There are still limitations, however, on the type of N-alkyl
structures that
are accepted by the cyclopamine target, according to our cell-based assays.
Compounds
with large steric bulk are unable to block Shh signaling at concentrations up
to 15 M
(derivatives 57, 58, and 63), either due to cell membrane impermeability or
steric
exclusion from the cyclopamine-interacting site. Even relatively small,
branched
elements close to the cyclopamine skeleton significantly abrogate biological
activity
(compound 50).

CA 02386190 2006-08-30
-98-
These compounds illustrate the variety of N-modifications that are accepted by
the cyclopamine target. It should be noted, however, that oxidation of the
hydroxyl
group to a ketone consistently improves the inhibitory activities of the
cyclopamine
derivatives by approximately two-fold (see compound 18 vs. 19; 20 vs. 21; 22
vs. 26; 33
vs. 34; 40 vs. 41; and 50 vs. 51). Formation of the enone by migration of the
C5-C6
olefin to the C4-C5 position (see compounds 26 and 41) does not appear to
affect
compound potency, suggesting that the importance of unsaturation at the C5-C6
position
reported by Keeler and co-workers is primarily due to sp2-hybridation at the
C5 carbon.
Collectively, these SAR data have facilitated the synthesis of potent Shh
signaling
inhibitors (for example, compound 34 is the most potent Shh inhibitor known to
date),
radiolabeled-probes (30 and 39), photoaffinity reagents (39 and 41), and
fluorophores
(43).
Most significantly, the compounds described in this study exemplify the
facility
by which derivatives of cyclopamine can be synthesized and evaluated. The
versatile
interrnediate 17 should promote the development of further cyclopamine-based
molecules, expediting the discovery of derivatives with desirable
pharmacological
properties. Such cyclopamine analogs could also be rapidly prepared through
combinatorial synthetic approaches; the cyclopamine skeleton of 17 could be
immobilized on a solid support via the C3-OH, and structurally diverse
functionalities
could be conjugated to the primary amine in a repetitive split-and-pool
manner. In
principle, this strategy would permit the simultaneous synthesis of millions
of
cyclopamine derivatives in a format amenable to high throughput screening.
Preparation of Compounds
General synthetic procedures. All reactions were performed under a positive
pressure of nitrogen. Air and moisture sensitive compounds were introduced via
syringe
or cannula through a rubber septum. All reagents and solvents were analytical
grade and
used as received with the following exceptions. DMF and DMSO were stored over
4 A
molecular sieves, and water was de-ionized and distilled. Flash chromatography
purifications were performed with the indicated solvent system on Merck silica
gel 60
(230-400 mesh). Low and high resolution mass spectra were obtained by the Mass
*Trade-mark

CA 02386190 2006-08-30
-99-
Spectrometry Facility at the Harvard Department of Chemistry and Chemical
Biology.
Proton magnetic resonance spectra (IH NMR) were recorded on a Varian 500 MHz
spectrophotometer.
H H
/ jI H tii
\p H
H H
H OH
1 2 3
HO H
Isolation of crude cyclopamine (1). A benzene extract of Veratrum
californicum (6.06 g) obtained from the United States Department of
Agriculture was
-10 purified by flash chromatography (Si02, step-wise gradient from 50:1 to
6.25:1
dichioromethane/ethanol to yield crude cyclopamine as a light brown solid (460
mg,
approximately 1.12 mmoles). 1H NMR: spectrum confirms the isolation of
cyclopamine
along with some impurities.
Isolation of crude jervine (2). A benzene extract of Veratruni virides (1.1 g)
obtained from the United States Department of Agriculture was purified by
flash
chromatography (Si02, step-wise gradient from 50:1 to 6.25:1
dichloromethane/ethanol)
to yield a brown oil (550 mg). The residue was then recrystallized in
ethanol/water (35
mL; 1:1) to yield a slightly yellow solid (215 mg, approximately 505 moles).
Concentration and recrystallization of the mother liquor yielded another batch
of crude
jervine (101 mg, approximately 237 moles) tH NMR: spectrum confirms the
isolation
ofjervine along with some impurities.
Cycloposine (3). Cycloposine was obtained from the United States Department
of Agriculture as a white solid.
*Trade-mark

CA 02386190 2002-04-11
WO 01/27135 PCT/US00/28479
- 100 -
4
Dihydrocinnamic acid N-hydroxysuccinimide ester (4). Dihydrocinnnamoyl
chloride (638 L, 4.21 mmoles) was added dropwise to a solution of N-
hydroxysuccinimide (500 mg, 4.21 mmoles) and triethylamine (704 L, 5.05
mmoles) in
dichloromethane (5 mL) at 0 C. The reaction was warmed to room temperature
stirred
for 1 h. The reaction mixture was then added to diethyl ether (50 mL), washed
with 1 N
HC1 (1 x 20 mL) and saturated aqueous NaHCO3 (1 x 20 mL), dried over MgSO4,
and
concentrated in vacuo to yield a white solid (1.03 g, 4.17 mmoles, 99%). HRMS:
(EI+)
= calcd for C13Ht3NO4 (M + H): 247.0845; found: 247.0841. 1H NMR: spectrum is
consistent with the predicted structure.
F3
Fi N ~q H
H
5
N-Trifluoroacetyl cyclopamine (5). Trifluoroacetic anhydride (77.3 L,
547 moles) was added to solution of crude cyclopamine (75.0 mg, 182 moles)
and
triethylamine (102 L, 729 moles) in dichloromethane (0.5 mL), and the
mixture was
stirred for 10 min at room temperature. The reaction mixture was evaporated to
dryness
by a stream of nitrogen gas and resuspended in MeOH (2 mL). The methanol
solution
was refluxed for 45 min then evaporated to dryness in vacuo. Purification by
flash
chromatography (Si02, step-wise gradient from 8:1 to 2:1 hexane:acetone)
yielded the
amide as a white solid (47.3 mg, 93.2 moles, 51%). LRMS: (ES+) calcd for
C29H40NO3F3 (M + H): 508; found: 508. 1H NMR: spectrum is consistent with the
predicted structure.

CA 02386190 2002-04-11
WO 01/27135 PCT/US00/28479
-101-
F3
F3
/ ~N
/ ~H
~ H
~ N ~
H ~ O
6
N-Trifluoroacetyl, 30-succinimidylcarbonyl cyclopamine (6). Disuccinimidyl
carbonate (107 mg, 417 moles) was added to solution of 5 (42.3 mg, 83.3
moles) and
triethylamine (116 L, 833 moles) in acetonitrile (1.0 mL), and the mixture
was stirred
5 for 5 h at room temperature. The reaction mixture was dissolved in diethyl
ether (10
mL), washed with 5% citric acid (1 x 2 mL) and saturated aqueous NaHCO3 (1 x 2
mL),
dried over MgSO4, and concentrated in vacuo. Purification by flash
chromatography
(Si02, step-wise gradient from 8:1 to 2:1 hexane/acetone) yielded the
carbonate as a
white solid (35.6 mg, 54.9 moles, 66%). LRMS: (ES+) calcd for C34H43N207F3 (M
+
H): 649; found: 649. 1H NMR: spectrum is consistent with the predicted
structure.
F3 F3
H
H \ I ~/\A O'C
FI
5ax -i
H
6 7
N-Trifluoroacetyl, 30-dihydrocinnamoylethylenediaminecarbamoyl
cyclopamine (7). Ethylenediamine (22.9 L, 342 moles) was added to solution
of 6
(11.1 mg, 17.1 moles) in dichloromethane (0.5 mL), and the mixture was
stirred for 15
min at room temperature. The reaction mixture was evaporated to dryness by a
stream of
nitrogen gas and excess ethylenediamine was removed in vacuo. The resultant
residue
was redissolved in dichloromethane (0.5 mL) and treated with 4 (10.6 mg, 4.28
moles)
and triethylamine (5.97 L, 42.8 moles). After stirring at room temperature
for 30 min,
the solution was filtered through a plug of glass wool. Purification by flash
chromatography (Si02, step-wise gradient from 4:1 to 1:1 hexane/acetone)
yielded the

CA 02386190 2002-04-11
WO 01/27135 PCT/US00/28479
- 102 -
carbamate as a white solid (6.7 mg, 9.23 moles, 54%). LRMS: (ES+) calcd for
C41H54N305F3 (M + H): 726; found: 726. 1H NMR: spectrum is consistent with the
predicted structure.
F3
H
/ I
fl H ~ H FI N
~ N/~ ~ ~ H\^ ~
H H
7 $
30-dihydrocinnamoylethylenediaminecarbamoyl cyclopamine (8).
Compound 7 (3.0 mg, 4.13 moles) was dissolved in a 2 M solution of ammonia in
methanol (0.5 mL, 1.00 mmoles). The reaction was stirred at room temperature
for 2 h
and then evaporated to dryness by a stream of nitrogen gas. Purification by
flash
chromatrography (Si02, step-wise gradient from 20:1:0.1 to 20:2:0.1
chloroform/methanol/triethylamine) yielded the amine as a colorless oil (2.1
mg, 3.33
moles, 81%). LRMS: (ES+) calcd for C39H55N304 (M + H): 630; found: 630. 1H
NMR: spectrum is consistent with the predicted structure.
F3
H
/ ~'i / ~
H p H
H ~ H \
2 9
N-Trifluoroacetyl jervine (9). Trifluoroacetic anhydride (84.4 L, 299 moles)
was added to solution of crude jervine (50.9 mg, 120 moles) and triethylamine
(100
L, 359 moles) in dichloromethane (0.5 mL), and the mixture was stirred for 15
min at
room temperature. The reaction mixture was evaporated to dryness by a stream
of
nitrogen gas and resuspended in MeOH (0.5 mL). The methanol solution was
stirred at
room temperature for 10 min then evaporated to dryness in vacuo. Purification
by flash
chromatography (Si02, step-wise gradient from 16:1 to 2:1 hexane:acetone)
yielded the

CA 02386190 2002-04-11
WO 01/27135 PCT/US00/28479
- 103-
amide as a white solid (37.8 mg, 72.5 moles, 60%). LRMS: (ES+) calcd for
C29H38N04F3 (M + H): 522; found: 522. 1H NMR: spectrum is consistent with the
predicted structure.
F3
Fs
-I
H
H \ O
9 10
N-Trifluoroacetyl, 30-succinimidylcarbonyl jervine (10). Disuccinimidyl
carbonate (92.9 mg, 363 moles) was added to solution of 9 (37.8 mg, 72.5
moles) and
triethylamine (101 L, 725 moles) in acetonitrile (1.0 mL), and the mixture
was stirred
for 6 h at room temperature. The reaction mixture was dissolved in diethyl
ether (10
mL), washed with 5% citric acid (1 x 2 mL) and saturated aqueous NaHCO3 (1 x 2
mL),
dried over MgSO4, and concentrated in vacuo. Purification by flash
chromatography
(Si02, step-wise gradient from 8:1 to 2:1 hexane/acetone) yielded the
carbonate as a
white solid (38.4 mg, 57.9 moles, 80%). HRMS: (ES+) calcd for C34H41N208F3 (M
+
Na): 685.2712; found: 685.2711. 1H NMR: spectrum is consistent with the
predicted
structure.
F3 F3
11 H
H fi
0 H
10 11
N-Trifluoroacetyl, 30-dihydrocinnamoylethylenediaminecarbamoyl jervine
(11). Ethylenediamine (20.2 L, 302 moles) was added to solution of 10 (10.0
mg,
15.1 moles) in dichloromethane (0.5 mL), and the mixture was stirred for 15
min at
room temperature. The reaction mixture was evaporated to dryness by a stream
of

CA 02386190 2002-04-11
WO 01/27135 PCT/US00/28479
- 104 -
nitrogen gas and excess ethylenediamine was removed in vacuo. The resultant
residue
was redissolved in dichloromethane (0.5 mL) and treated with 4 (7.47 mg, 30.2
moles)
and triethylamine (4.21 L, 30.2 moles) at 0 C. After stirring at 0 C for 30
min, the
solution was filtered through a plug of glass wool. Purification by flash
chromatography
(SiOZ, step-wise gradient from 4:1 to 1:1 hexane/acetone) yielded the
carbamate as a
white solid (8.0 mg, 10.8 moles, 72%). LRMS: (ES+) calcd for C41H52N306F3 (M
+
H): 740; found: 740. 1H NMR: spectrum is consistent with the predicted
structure.
F3
H
/ ~ / ~
H FI H H ~{ H
~ ~~ ~
H H
11 12
30-dihydrocinnamoylethylenediaminecarbamoyl jervine (12). Aqueous
ammonia (200 L of a 29% (w/w) solution in water, 3.04 mmoles) was added to a
solution of 11 (1.0 mg, 1.35 moles) in methanol (200 L). The reaction was
stirred at
room temperature for 30 min and then evaporated to dryness by a stream of
nitrogen gas.
Purification by flash chromatrography (Si02, step-wise gradient from 20:1:0.1
to
20:2:0.1 chloroform/methanol/triethylamine) yielded the amine as a colorless
oil (0.8
mg, 1.24 moles, 92%). LRMS: (ES+) calcd for C39H53N305 (M + H): 644; found:
644. 1H NMR: spectrum is consistent with the predicted structure.
HZHY H ~ F3c H~H
13
N-Trifluoroacetyl glycine (13). Methyl trifluoroacetate (804 L, 7.99 mmoles)
and triethylamine (928 L, 6.66 mmoles) were added to a suspension of glycine
(500
mg, 6.66 mmoles) in methanol (2.5 mL). After the mixture was stirred
vigorously for 18
h, 1 N HC1 was added dropwise until the a pH of 2 was obtained. The reaction
was
added to ethyl acetate (30 mL) was washed with 1 N HCI (2 x 10 mL), dried over
MgSO4, and concentrated in vacuo to yield the amide as a white solid (991 mg,
5.79

CA 02386190 2002-04-11
WO 01/27135 PCT/US00/28479
-105-
mmoles, 87%). LRMS: (CI+) calcd for C4H4NO3F3 (M + NH4): 189; found: 189. 1H
NMR: spectrum is consistent with the predicted structure.
F3AHWIIH AHN'~
V~- 13(~ 14
N-Trifluoroacetyl glycine N-hydrosuccinimide ester (14). Disuccinimidyl
carbonate (300 mg, 1.17 mmoles) was added to a solution of 13 (200 mg, 1.17
mmoles)
and pyridine (94.6 L, 1.17 mmoles) in acetonitrile (1.0 mL). The reaction
mixture was
stirred at room temperature for 3 h, during which the solution became clear
and evolved
gas. The solution was added to ethyl acetate (10 mL), washed with 1N HCI (2 x
5 mL)
and saturated aqueous NaHCO3 (2 x 5 mL), dried over MgSO4, and concentrated in
vacuo to yield a white solid (232 mg, 865 moles, 74%). LRMS: (ES+) calcd for
C8H7N205F3 (M + NH4): 286; found: 286. 1H NMR: spectrum is consistent with the
predicted structure.
H
CF3
H
/ 11 / 1-I
~ H ~ FI
H \ H ~
1 15
N-(N'-Trifluoroacetyl glycyl) cyclopamine (15). Triethylamine (135 L,
972 moles) and 14 (261 mg, 972 moles) were added to a solution of
cyclopamine in
dichloromethane (2.0 mL). The reaction was stirred at room temperature for 1 h
and then
subjected directly to purification by flash chromatrography (Si02, step-wise
gradient
from 8:1 to 2:1 hexane/acetone) yielded the amide as a white solid (166 mg,
294 moles,
60%). LRMS: (ES+) calcd for C31H43N204F3 (M + H): 565; found: 565. 1H NMR:
spectrum is consistent with the predicted structure.

CA 02386190 2002-04-11
WO 01/27135 PCT/US00/28479
- 106 -
~N~F3 NHZ
/ H / ~H
1I FI H
H ~ H \
15 16
N-Glycyl cyclopamine (16). Aqueous ammonia (3 mL of a 29% (w/w) solution
in water, 45.6 mmoles) was added to a solution of 15 (162 mg, 296 moles) in
methanol
(4 mL). The reaction was stirred at room temperature for 5 h and then
evaporated to
dryness in vacuo. Purification by flash chromatrography (Si02; chloroform,
then step-
wise gradient from 20:1:0.1 to 20:2:0.1 chloroform/methanol/triethylamine)
yielded the
amine as a white solid (110 mg, 235 moles, 79%). LRMS: (ES+) calcd for
C29H44N203 (M + H): 469; found: 469. 1H NMR: spectrum is consistent with the
predicted structure.
~tJHp lNH2
/ H / i~
~{ FI ~{ H
H \ H \
16 17
N-Aminoethyl cyclopamine (17). Lithium aluminum hydride (939 gL of a 1 M
solution in THF, 939 moles) was added to a suspension of 16 (110 mg, 235
moles) in
THF (6 mL). The reaction was refluxed for 3 h and then quenched with water (5
mL)
and aqueous KOH (10 mL of a 10% solution). After extracting the mixture with
chloroform (2 x 20 mL), the organic layer was dried over Na2SO4, filtered, and
concentrated in vacuo. Purification by flash chromatography (Si02, step-wise
gradient
from 20:1:0.1 to 20:2:0.1 chloroform/methanol/triethylamine) yielded the
diamine as a
colorless oil (94.4 mg, 208 moles, 88%). LRMS: (ES+) calcd for C29H46N202 (M
+
H): 455; found: 455. 1H NMR: spectrum is consistent with the predicted
structure.

CA 02386190 2002-04-11
WO 01/27135 PCT/US00/28479
- 107 -
~
CNHZ H \ I
/ H / ~H
w H ~ H
H \ H \
17 18
N-(N'-Dihydrocinnamoyl aminoethyl) cyclopamine (18). Triethylamine
(5.03 L, 36.1 moles) and 4 (4.46 mg, 18.0 moles) were added to a solution
of 17 (8.2
mg, 18.0 moles) in dichloromethane (500 L). The reaction was stirred at room
temperature for 3 h and then evaporated to dryness by a stream of nitrogen
gas.
Purification by flash chromatography (Si02, step-wise gradient from 4:1 to 1:1
hexane/acetone) yielded the amide as a white solid (5.7 mg, 9.71 moles, 54%).
LRMS:
(ES+) calcd for C38H54N203 (M + H): 587; found: 587. 1H NMR: spectrum is
consistent with the predicted structure.
H H
O 0
/ H , H
\ H A H
H
18 19
3-Keto, N-(N'-dihydrocinnamoyl aminoethyl) cyclopamine (19).
Dimethylsulfoxide (6.89 L, 97.1 moles) was added to a solution of oxalyl
chloride
(4.24 L, 48.6 moles) in dichloromethane (250 L) at -78 C. After the
mixture was
stirred at -78 C for 10 min, a solution of 18 (5.7 mg, 9.71 moles) in
dichloromethane
(250 L) was added, and the reaction was stirred at -78 C for another 30 min.
The
oxidation was completed by the addition of triethylamine (20.3 L, 146 moles)
to the
solution, which was stirred at -78 C for 10 min and then allowed to warm to
room
temperature. The reaction was quenched by the addition of water (1 mL) and
chloroform
(5 mL), and the organic layer was isolated, washed with brine (1 x 2 mL),
dried over
over Na2SO4, and concentrated in vacuo. Purification by flash chromatography
(Si02,

CA 02386190 2002-04-11
WO 01/27135 PCT/US00/28479
-108-
step-wise gradient from 8:1 to 4:1 hexane/acetone) to yielded the ketone as a
white solid
(2.0 mg, 3.42 gmoles, 35%). Recovered starting material (1.6 mg, 2.73 gmoles,
28%).
LRMS: (ES+) calcd for C38H52N203 (M + H): 585; found: 585. 1H NMR: spectrum is
consistent with the predicted structure.
~NHZ H
/ i-I / H
\ FI \ H
H H
17 20
N-(N'-(4-Benzoylbenzoyl) aminoethyl) benzophenone (20). 4-Benzoylbenzoic
acid N-hydroxysuccinimide ester (8.01 mg, 23.5 gmoles) and triethylamine (6.55
gL,
47.0 moles) were added to a solution of 17 (10.7 mg, 23.5 gmoles) in
dichloromethane
(500 gL). The reaction was stirred at room temperature for 2 h and then
evaporated to
dryness by a stream of nitrogen gas. Purification by flash chromatography
(Si02, step-
wise gradient from 100:1 to 50:1 chloroform/methanol) yielded the benzophenone
as a
colorless oil (10.8 mg, 16.3 gmoles, 69%). LRMS: (ES+) calcd for C43H54N204 (M
+
H): 663; found: 663. 1H NMR: spectrum is consistent with the predicted
structure.
H H ~I I,
~ o
H
N H
H
21
3-Keto, N-(N'-(4-benzoylbenzoyl) aminoethyl) cyclopamine (21).
Dimethylsulfoxide
(11.6 gL, 163 gmoles) was added to a solution of oxalyl chloride (7.11 gL,
81.5 gmoles)
in dichloromethane (250 gL) at -78 C. After the mixture was stirred at -78 C
for 10
20 min, a solution of 20 (10.8 mg, 16.3 moles) in dichloromethane (250 gL)
was added,

CA 02386190 2002-04-11
WO 01/27135 PCT/US00/28479
- 109 -
and the reaction was stirred at -78 C for another 30 min. The oxidation was
completed
by the addition of triethylamine (34.1 L, 245 moles) to the solution, which
was stirred
at -78 C for 10 min and then allowed to warm to room temperature. The reaction
was
quenched by the addition of water (1 mL) and chloroform (5 mL). The resultant
organic
layer was then washed with brine (1 x 2 mL), dried over over Na2SO4, and
concentrated
in vacuo. Purification by flash chromatography (Si02, step-wise gradient from
8:1 to 2:1
hexane/acetone) yielded the ketone as a white solid (6.3 mg, 9.53 moles,
58%). LRMS:
(ES+) calcd for C43H52N204 (M + H): 661; found: 661. 1H NMR: spectrum is
consistent with the predicted structure.
CNHZ H _N
H
H H
17 22
N-(N'-Azidoiodophenylpropionyl aminoethyl) cyclopamine (22).
Azidoiodophenylpropionyl N-hydroxysuccinimide ester (1.9 mg, 4.18 moles) and
triethylamine (2.34 L,16.7 moles) were added to a solution of 17 (1.9 mg,
4.18
moles) in dichloromethane (500 L). The reaction was stirred at room
temperature for
3 h and then evaporated to dryness by a stream of nitrogen gas. Purification
by flash
chromatography (Si02, step-wise gradient from 4:1 to 1:1 hexane/acetone)
yielded the
benzophenone as a white solid (1.6 mg, 2.12 moles, 51%). LRMS: (ES+) calcd
for
C38H52N5031 (M + H): 754; found: 754. IH NMR: spectrum is consistent with the
predicted structure.

CA 02386190 2002-04-11
WO 01/27135 PCT/US00/28479
- 110 -
H
~NHZ itJ\ /CF3
/\ n0
/ H
H
FI ~{ H
H H
17 23
N-(N'-Trifluoroacetyl aminoethyl) cyclopamine (23). Trifluoroacetic
anhydride (20.6 L, 146 moles) and triethylamine (24.5 L, 176 moles) were
added
to a solution of 17 (13.3 mg, 29.3 moles) in dichloromethane (0.5 mL). The
mixture
was stirred for 30 min at room temperature and then evaporated to dryness by a
stream
of nitrogen gas The resultant residue was redissolved in methanol (1 mL) and
the
solution was stirred at room temperature for 1 h. After removal of the solvent
in vacuo,
purification by flash chromatography (Si02, step-wise gradient from 4:1 to 2:1
hexane:acetone) yielded the amide as a white solid (9.2 mg, 16.7 moles, 57%).
LRMS:
not performed. 1H NMR: spectrum is consistent with the predicted structure.
H CF3 ~CF3
\ p O
/ H / H
~ li ry H
H \ O ~
23 24
3-Keto, N-(N'-trifluoroacetyl aminoethyl) cyclopamine (24).
Dimethylsulfoxide (11.9 jiL, 167 moles) was added to a solution of oxalyl
chloride
(7.28 L, 83.5 moles) in dichloromethane (250 L) at -78 C. After the
mixture was
stirred at -78 C for 10 min, a solution of 23 (9.2 mg, 16.7 moles) in
dichloromethane
(250 L) was added, and the reaction was stirred at -78 C for another 30 min.
The
oxidation was completed by the addition of triethylamine (35.0 L, 251 moles)
to the
solution, which was stirred at -78 C for 10 min and then allowed to warm to
room
temperature. The reaction was quenched by the addition of saturated aqueous
NaHCO3
(2 mL) and chloroform (5 mL), and the organic layer was isolated, dried over
Na2SO4,
and concentrated in vacuo. Purification by flash chromatography (Si02, step-
wise

CA 02386190 2002-04-11
WO 01/27135 PCT/US00/28479
- 111 -
gradient from 8:1 to 4:1 hexane/acetone) yielded the ketone as a white solid
(6.0 mg,
10.9 moles, 65%). LRMS: not performed. 1H NMR: spectrum is consistent with
the
predicted structure.
H
ifJfCF3 (-NH2
/ H / H
~ ~ ~ ~y H
~
24 25
3-Enone, N-aminoethyl cyclopamine (25). Aqueous ammonia (250 L of a
29% (w/w) solution in water, 3.80 mmoles) was added to a solution of 24 (3.0
mg, 5.47
moles) in methanol (250 L). The reaction was stirred at room temperature for
24 h
and then evaporated to dryness by a stream of nitrogen gas. Purification by
flash
chromatrography (Si02, step-wise gradient from 20:1:0.1 to 20:2:0.1
chloroform/methanol/triethylamine) yielded the amine as a colorless oil (3.0
mg, 6.63
moles, quant.). LRMS: not performed. 1H NMR: spectrum is consistent with the
predicted structure.
N
H2 H ~ I N
O
/ H / FI
H FI ~ H
i /
25 26
3-Enone, N-(N'-azidoiodophenylpropionyl aminoethyl) cyclopamine (26).
Azidoiodophenylpropionyl N-hydroxysuccinimide ester (1.4 mg, 3.31 moles) and
triethylamine (1.8 L, 13.2 moles) were added to a solution of 25 (1.5 mg,
3.31
moles) in dichloromethane (250 L). The reaction was stirred at room
temperature for
3 h and then quenched with dimethylaminopropylamine. Purification by flash
chromatography (Si02, step-wise gradient from 8:1 to 2:1 hexane/acetone) to
yield the

CA 02386190 2002-04-11
WO 01/27135 PCT/US00/28479
- 112 -
aryl azide as a white solid (0.5 mg, 0.665 moles, 20%). LRMS: (ES+) calcd for
C38H50N503I (M + H): 752; found: 752. 1H NMR: spectrum is consistent with the
predicted structure.
Hp H F3~ry H
27
N-Trifluoroacetyl 12-aminododecanoic acid (27). Methyl trifluoroacetate (200
L, 1.99 mmoles) and triethylamine (184 L, 1.32 mmoles) were added to a
suspension
of 12-aminododecanoic acid (300 mg, 1.32 mmoles) in methanol (2 mL). After the
mixture was stirred vigorously for 18 h, 1 N HCl was added dropwise until the
a pH of 2
was obtained. The reaction was added to ethyl acetate (20 mL) was washed with
1 N
HCl (2 x 5 mL), dried over MgSO4, and concentrated in vacuo to yield the amide
as a
white solid (398 mg, 1.28 mmoles, 97%). LRMS: not performed. 1H NMR: spectrum
is
consistent with the predicted structure.
F3C H H FsAH
27 28
N-Trifluoroacetyl 12-aminododecanoic acid N-hydroxysuccinimide ester
(28). Disuccinimidyl carbonate (247 mg, 964 gmoles) was added to a solution of
27
(200 mg, 642 moles) and pyridine (104 L, 1.28 mmoles) in acetonitrile (2.0
mL). The
reaction mixture was stirred at room temperature for 4.5 h, during which the
solution
became clear and evolved gas. The solution was added to ethyl acetate (10 mL),
washed
with 1 N HCl (2 x 1 mL) and saturated aqueous NaHCO3 (2 x 1 mL), dried over
MgSO4, and concentrated in vacuo to yield a white solid (257 mg, 629 moles,
98%).
LRMS: not performed. 1H NMR: spectrum is consistent with the predicted
structure.

CA 02386190 2002-04-11
WO 01/27135 PCT/US00/28479
-113-
H
NHZ CF3
H
/ '~ / .~
~{ FI ~ H
H \ H \
17 29
N-(N'-(N"-Trifluoroacetyl aminododecanoyl) aminoethyl) cyclopamine
(29).Triethylamine (4.78 L, 34.3 moles) and 28 (8.4 mg, 20.6 moles) were
added to
a solution of 17 (7.8 mg, 17.2 moles) in dichloromethane (250 L). The
reaction was
stirred at room temperature for 12 h and then evaporated to dryness by a
stream of
nitrogen gas. Purification by flash chromatography (Si02, step-wise gradient
from 100:1
to 25:1 chloroform/methanol) yielded the amide as a white solid (8.0 mg, 10.7
moles,
62%). LRMS: (ES+) calcd for C43H68N304 (M + H): 748; found: 748. 1H NMR:
spectrum is consistent with the predicted structure.
H
NHZ
/ ~ / H
p H ~y H
H \ H \
17 30
N-(N'-Propionyl aminoethyl) cyclopamine (30). Propionyl
N-hydroxysuccinimide ester (1.08 mg, 6.33 moles) and triethylamine (1.48 L,
10.6
moles) were added to a solution of 17 (2.4 mg, 5.28 moles) in dichloromethane
(250
L). The reaction was stirred at room temperature for 12 h and then quenched
with
dimethylaminopropylamine. Purification by flash chromatography (Si02, step-
wise
gradient from 100:1 to 25:1 chloroform/methanol) yielded the amide as a
colorless oil
(1.8 mg, 3.52 moles, 67%). LRMS: (ES+) calcd for C32H50N203 (M + H): 511;
found: 511. 1H NMR: spectrum is consistent with the predicted structure.
Preparation of 3H-labeled 30. Propionyl N-hydroxysuccinimide ester (1 mCi,
specific activity = 100 Ci/mmol, 10 nmoles) in ethyl acetate (1.0 mL) was
mixed with 17
(1.1 mg, 2.5 moles) in chloroform (100 L). The reaction mixture was
incubated

CA 02386190 2002-04-11
WO 01/27135 PCT/US00/28479
- 114 -
without stirring for 20 h at room temperature. Purification by flash
chromatography
(Si02, step-wise gradient from 100:1 to 25:1 chloroform/methanol) yielded the
tritium-
labeled cyclopamine derivative. Fractions containing the desired product were
pooled
and concentrated with a stream of nitrogen gas. The concentration solution was
resuspended in methanol (200 L) and stored at -20 C. Beta/scintillation
counter
analysis determined the reaction yield to be approximately 81%, and thin layer
chromatography analysis (Rf = 0.80; 10:2:0.1
dichloromethane/methanol/triethylamine)
is consistent with known properties of cold 30.
HZN-~ " H
31
N-Dihydrocinnamoyl aminocaproic acid (31). Aminocaproic acid (100 mg,
747 moles) and 4 (185 mg, 747 moles) were dissolved in DMF/water (1 mL;
1:1).
The reaction was stirred at room temperature for 1 h, and then acidified with
1 N HCl
until a pH of 2 was obtained. The mixture was added to ethyl acetate (10 mL),
washed
with 1 N HCl (2 x 5 mL), dried over MgSO4, and concentrated in vacuo to yield
a white
waxy solid (175 mg, 665 moles, 89%). LRMS: not performed. 1H NMR: spectrum is
consistent with the predicted structure.
OH ~ ~
O
C",Q
31 32
N-Dihydrocinnamoyl aminocaproic acid N-hydroxysuccinimide ester (32).
Disuccinimidyl carbonate (155 mg, 604 moles) was added to a solution of 31
(159 mg,
604 moles) and pyridine (97.7 L, 1.21 mmoles) in acetonitrile (1 mL). The
reaction
mixture was stirred at room temperature for 2.5 h, during which the solution
became
clear and evolved gas. The solution was added to ethyl acetate (5 mL), washed
with 1 N
HCl (2 x 1 mL) and saturated aqueous NaHCO3 (2 x 1 mL), dried over MgSO4, and
concentrated in vacuo to yield a colorless oil (156 mg, 433 moles, 72%).
LRMS: not
performed. 1H NMR: spectrum is consistent with the predicted structure.

CA 02386190 2002-04-11
WO 01/27135 PCT/US00/28479
-115-
NHy H
C"~H
/ H
~ H
H H
17 33
N-(N'-(N"-Dihydrocinnamoyl aminocaproyl) aminoethyl) cyclopamine (33).
Triethylamine (4.43 L, 31.8 moles) and 32 (5.75 mg, 15.9 moles) were added
a
solution of 17 (7.25 mg, 15.9 moles) in dichloromethane (250 L). The
reaction was
stirred at room temperature for 1 h and evaporated to dryness by a stream of
nitrogen
gas. Purification by flash chromatography (Si02, step-wise gradient from 100:1
to 25:1
chloroform/methanol) yielded the amide as a colorless oil (5.8 mg, 8.29
moles, 52%).
LRMS: (ES+) calcd for C44H65N304 (M + H): 700; found: 700. 1H NMR: spectrum is
consistent with the predicted structure.
H H
A
lOl 0 H
~
~
~{ H ~ H
H \ \
33 34
3-Keto, N-(N'-(N"-dihydrocinnamoyl aminocaproyl) aminoethyl)
cyclopamine (34). Dimethylsulfoxide (12.7 L, 177 moles) was added to a
solution of
oxalyl chloride (7.73 L, 88.6 moles) in dichloromethane (250 L) at -78 C.
After the
mixture was stirred at -78 C for 10 min, a solution of 33 (6.2 mg, 8.86
moles) in
dichloromethane (250 L) was added, and the reaction was stirred at -78 C for
another
30 min. The oxidation was completed by the addition of triethylamine (37.1 L,
266
moles) to the solution, which was stirred at -78 C for 10 min and then
allowed to
warm to room temperature. The reaction was quenched by the addition of
saturated
aqueous NaHCO3 (2 mL) and extracted with chloroform (2 x 2 mL). The resultant
organic layer was then isolated, dried over over Na2SO4, and concentrated in
vacuo.

CA 02386190 2002-04-11
WO 01/27135 PCT/USOO/28479
- 116 -
Purification by flash chromatography (Si02, step-wise gradient from 100:1 to
25:1
chloroform/methanol) to yielded the ketone as a slightly yellow oil (5.4 mg,
7.74
moles, 87%). LRMS: (ES+) calcd for C44H63N304 (M + H): 698; found: 698. 1H
NMR: spectrum is consistent with the predicted structure.
H2 H F3AH~ ~ ^ ^ H
35 _ fCnjS
N-Trifluoroacetyl aminocaproic acid (35). Methyl trifluoroacetate (513 L,
5.10 mmoles) and triethylamine (474 L, 3.40 mmoles) were added to a
suspension of
aminocaproic acid (455 mg, 3.40 mmoles) in methanol (2 mL). After the mixture
was
stirred vigorously for 8 h, 1 N HCI was added dropwise until the a pH of 2 was
obtained.
The reaction was added to ethyl acetate (10 mL) was washed with 1 N HCI (2 x 2
mL),
dried over MgSO4, and concentrated in vacuo to yield the amide as a white
solid (745
mg, 3.49 mmoles, quant.). LRMS: not performed. 1H NMR: spectrum is consistent
with
the predicted structure.
H F3~
F3~H H
35 i5 36
N-Trifluoroacetyl aminocaproic acid N-hydroxysuccinimide ester (36).
Disuccinimidyl carbonate (541 mg, 2.11 mmoles) was added to a solution of 35
(300
mg, 1.41 mmoles) and pyridine (227 L, 2.81 mmoles) in acetonitrile (2.0 mL).
The
reaction mixture was stirred at room temperature for 13 h, during which the
solution
became clear and evolved gas. The solution was added to ethyl acetate (10 mL),
washed
with 1 N HCl (2 x 1 mL) and saturated aqueous NaHCO3 (2 x 1mL), dried over
MgSO4,
and concentrated in vacuo to yield a white solid (471 mg, 1.45 mmoles,
quant.). LRMS:
not performed. 1 H NMR: spectrum is consistent with the predicted structure.

CA 02386190 2002-04-11
WO 01/27135 PCT/US00/28479
-117-
H
CNHZ ~NH CF3
/ jI / ~ H H \ H 17 37
N-(N'-(N"-Trifluoroacetyl aminocaproyl) aminoethyl) cyclopamine (37).
Triethylamine (12.3 L, 88.0 moles) and 36 (17.1 mg, 52.8 moles) were added
to a
solution of 17 (20.0 mg, 44.0 moles) in dichloromethane (200 L). The
reaction
mixture was stirred at room temperature for 13 h, quenched with
dimethylaminopropylamine (11.2 L, 88.0 moles), and evaporated to dryness
with a
stream of nitrogen gas. Purification by flash chromatography (Si02, step-wise
gradient
from 50:1 to 25:1 chloroform/methanol) yielded the amide as a colorless oil
(26.9 mg,
40.5 moles, 92%). LRMS: not performed. 1H NMR: spectrum is consistent with
the
predicted structure.
N H
~ --- CF3 NHp
H \ ~~55
/ N / ~
~y FI (y H
H \ H \
37 38
N-(N'-Aminocaproyl aminoethyl) cyclopamine (38). Aqueous ammonia (200
L of a 29% (w/w) solution in water, 3.04 mmoles) was added to a solution of 37
(26.9
mg, 40.5 moles) in methanol (400 L). The reaction was stirred at room
temperature
for 20 h and then evaporated to dryness by a stream of nitrogen gas.
Purification by flash
chromatrography (Si02, step-wise gradient from 20:1:0.1 to 20:4:0.1
chloroform/methanol/triethylamine) yielded the amine as a white waxy solid
(19.3 mg,
34.0 moles, 84%.). LRMS: not performed. 1H NMR: spectrum is consistent with
the
predicted structure.

CA 02386190 2002-04-11
WO 01/27135 PCT/US00/28479
- 118 -
H H
C I
NHZ H I
/ H
~ ~ (~
H ~
H
38 39
N-(N'-(N"-Azidoiodophenylpropionyl aminocaproyl) aminoethyl)
cyclopamine (39). Azidoiodophenylpropionyl N-hydroxysuccinimide ester (1.4 mg,
3.38 gmoles) and triethylamine (0.94 gL, 6.76 gmoles) were added to a solution
of 38
(1.92 mg, 3.38 moles) in dichloromethane (250 gL). The reaction was stirred
at room
temperature for 2.5 h and evaporated to dryness by a stream of nitrogen gas.
Purification
by flash chromatography (Si02, step-wise gradient from 4:1:0.025 to 1:2:0.015
hexane/acetone/triethylamine) yielded the azide as a colorless oil (2.2 mg,
2.54 gmoles,
75%). LRMS: (ES+) calcd for C44H63N604I (M + H): 867; found: 867. 1H NMR:
spectrum is consistent with the predicted structure.
Preparation of 1251-labeled 39. 125I-labeled azidoiodophenylpropionyl N-
hydroxysuccinimide ester (0.250 mCi, specific activity = 2200 Ci/mmol, 0.114
nmoles)
in ethyl acetate (2.1 mL) was concentrated to a volume of approximately 10 gL
by a
stream of nitrogen gas. The concentrated solution was diluted with ethyl
acetate (100
L) and was mixed with 38 (1.0 mg, 1.76 gmoles) in chloroform (100 L). The
reaction
was mixture was incubated without stirring for 43 h at room temperature and
then
concentrated to approximately 10 L by a stream of nitrogen gas. The residue
was
resuspended in chloroform (200 gL) and purified by flash chromatography (Si02,
step-
wise gradient from 100:1 to 12.5:1 chloroform/methanol) to yield the
radiolabeled azide.
Fractions containing the desired product were pooled, concentrated by a stream
of
nitrogen gas, resuspended in methanol (1 mL), and a small aliquot removed for
quantitation. Gamma counter analysis determined the reaction yield to be
essentially
quantitative, and the solution was reconcentrated by a stream of nitrogen gas,
resuspended in methanol (250 gL), and stored at -20 C. Thin layer
chromatography
analysis (Rf = 0.62; 10:2:0.1 dichloromethane/methanol/triethylamine) is
consistent with
known properties of cold 39.

CA 02386190 2002-04-11
WO 01/27135 PCT/US00/28479
- 119 -
H H
~.y ~ ~~ H H=...
C ?f ~ ~ NHZ H
la CH
}I
R N F
H
38 40
N-(N'-(N"-Biotinoyl aminocaproyl) aminoethyl) cyclopamine (40). Biotinoyl
N-hydroxysuccinimide ester (6.72 mg, 14.8 moles) and triethylamine (3.43 L,
24.6 moles) were added to a solution of 38 (5.6 mg, 12.3 moles) in DMF (250
L).
The reaction was stirred at room temperature for 14 h and then added to
chloroform (2
mL). The organic mixture was washed with saturated aqueous NaHCO3 (3 x 1 mL),
dried over Na2SO4, and concentrated in vacuo. Purification by flash
chromatography
(Si02, step-wise gradient from 20:1:0.05 to 20:5:0.05
chloroform/methanol/triethylamine) yielded the ketone as a white solid (9.4
mg, 12.5
moles, quant.). LRMS: (ES+) calcd for C45H71N505S (M + H): 794; found: 794. 1H
NMR: spectrum is consistent with the predicted structure.
H H NH H H NH
C O H ~ H
/ ~1-I / ~1-I
N ~ H
~Fi
H
40 41
3-Enone, N-(N'-(N"-Biotinoyl aminocaproyl) aminoethyl) cyclopamine (41).
Dimethylsulfoxide (3.66 L, 51.6 moles) was added to a solution of oxalyl
chloride
(2.25 L, 25.8 moles) in dichloromethane (250 L) at -78 C. After the
mixture was
stirred at -78 C for 10 min, a solution of 40 (4.1 mg, 5.16 moles) in
dichloromethane
(200 L) was added, and the reaction was stirred at -78 C for another 30 min.
The
oxidation was completed by the addition of triethylamine (10.8 L, 77.4
moles) to the
solution, which was stirred at -78 C for 10 min and then allowed to warm to
room

CA 02386190 2002-04-11
WO 01/27135 PCT/US00/28479
- 120 -
temperature. The reaction was quenched by the addition of saturated aqueous
NaHCO3
(2 mL) and extracted with chloroform (2 x 2 mL). The resultant organic layer
was then
dried over over Na2SO4, and concentrated in vacuo. The residue was redissolved
in
MeOH (0.5 mL) and treated with aqueous ammonia (250 L of a 29% (w/w) solution
in
water, 3.80 mmoles) for 20 h at room temperature. The reaction was added to
chloroform (2 mL), washed with saturated aqueous NaHCO3 (2 x 2 mL), dried over
Na2SO4, and concentrated in vacuo. Purification by flash chromatography (Si02,
step-
wise gradient from 20:1:0.1 to 20:5:0.1 chloroform/methanol/triethylamine) to
yielded
the enone as a yellowish solid (1.4 mg, 1.77 moles, 34%). LRMS: (ES+) calcd
for
C45H69N505S (M + H): 792; found: 792. 1H NMR: spectrum is consistent with the
predicted structure.
H
CN~NHZ H
IHI
}~
/
Fi ~ N
H ~
H
38 42
N-(N'-(N"-Propionyl aminocaproyl) aminoethyl) cyclopamine (42).
Propionyl N-hydroxysuccinimide ester (1.00 mg, 5.87 moles) and triethylamine
(1.37
L, 9.80 moles) were added to a solution of 38 (2.78 mg, 4.90 moles) in
dichloromethane (250 L). The reaction was stirred at room temperature for 12
h and
then quenched with dimethylaminopropylamine. Purification by flash
chromatography
(Si02, step-wise gradient from 100:1 to 10:1 chlorofonn/methanol) yielded the
amide as
a colorless oil (2.5 mg, 4.01 moles, 82%). LRMS: (ES+) calcd for C38H61N304
(M +
H): 624; found: 624. 1H NMR: spectrum is consistent with the predicted
structure.

CA 02386190 2002-04-11
WO 01/27135 PCT/US00/28479
- 121 -
H
(N_r~~ NHy H
~~
~ H
~I H 'Ir
H H
38 43
N-(N'-(N"-BODIPY FL aminocaproyl) aminoethyl) cyclopamine (43).
BODIPY FL N-hydroxysuccinimide ester (2.0 mg, 5.28 moles) and triethylamine
(0.98
L, 7.04 moles) were added to a solution of 38 (2.0 mg, 3.52 moles) in
dichloromethane (500 L). The reaction was stirred at room temperature for 20
h and
then evaporated to dryness with a stream of nitrogen gas. Purification by
flash
chromatography (Si02, step-wise gradient from 50:1 to 12.5:1
chloroform/methanol)
yielded the fluorophore as a colorless oil (2.6 mg, 3.09 moles, 88%). LRMS:
(ES+)
calcd for C49H70N504BF2 (M + H): 842; found: 842. 1H NMR: spectrum is
consistent
with the predicted structure.
~I I~ ~I I~
H
HZ F3~ H
H
N-Trifluoroacetyl 4-benzoylphenylalanine (45). Methyl trifluoroacetate (89.6
15 L, 891 moles) and triethylamine (104 L, 743 moles) were added to a
suspension of
4-benzoylphenylalanine (200 mg, 743 moles) in methanol (0.5 mL). The reaction
was
stirred vigorously for 24 h and then acidified with 1 N HCl until a pH of 2
was obtained.
The mixture was added to ethyl acetate (10 mL), washed with 1 N HCI (2 x 10
mL),
dried over MgSO4, and concentrated in vacuo to yield a white solid (49.7 mg,
136
20 moles, 18%). LRMS: not performed. IH NMR: spectrum is consistent with the
predicted structure.

CA 02386190 2002-04-11
WO 01/27135 PCT/US00/28479
-122-
~~ ~~ ~~ ~~
H
F3 H F3
H
N-Trifluoroacetyl 4-benzoylphenylalanine N-hydroxysuccinimide ester (46).
Disuccinimidyl carbonate (33.0 mg, 129 moles) was added to a solution of 45
(47.1
mg, 129 moles) and pyridine (20.9 L, 258 moles) in acetonitrile (0.5 mL).
The
5 reaction mixture was stirred at room temperature for 3 h, during which the
solution
became clear and evolved gas. The solution was added to ethyl acetate (5 mL),
washed
with 1 N HC1 (2 x 1 mL) and saturated aqueous NaHCO3 (2 x 1 mL), dried over
MgSO4, and concentrated in vacuo to yield a white solid (48.8 mg, 106 moles,
82%).
LRMS: not perfonned. 1H NMR: spectrum is consistent with the predicted
structure.
H
47
Levulinic acid N-hydroxysuccinimide ester (47). Disuccinimidyl carbonate
(692 mg, 2.70 mmoles) and pyridine (437 L, 2.70 mmoles) was added to a
solution of
levulinic acid (320 mg, 2.70 mmoles) in acetonitrile (2.0 mL). The reaction
mixture was
stirred at room temperature for 4.5 h, during which the solution became clear
and
evolved gas. The solution was added to ethyl acetate (10 mL), washed with 1 N
HC1(2 x
5 mL) and saturated aqueous NaHCO3 (2 x 5 mL), dried over MgSO4, and
concentrated
in vacuo to yield a white solid (333 mg, 1.56 mmoles, 58%). LRMS: not
performed. 1H
NMR: spectrum is consistent with the predicted structure.

CA 02386190 2002-04-11
WO 01/27135 PCT/US00/28479
-123-
/~
~ ~/
~
~H2 ACF3
/ ~
~ N ~ N
H H
17 48
N-(N'-(N"-Trifluoroacetyl 4-benzoylphenylalanine) aminoethyl)
cyclopamine (48). Triethylamine (12.6 L, 90.6 moles) and 46 (20.9 mg, 45.3
moles)
were added to a solution of 17 (20.6 mg, 45.3 moles) in dichloromethane (0.5
mL). The
reaction was stirred at room temperature for lh and then evaporated to dryness
by a
stream of nitrogen gas. Purification by flash chromatography (Si02, step-wise
gradient
from 8:1 to 1:1 hexane/acetone) yielded the benzephenone as a white solid
(22.7 mg,
28.3 moles, 62%). LRMS: not performed. 1H NMR: spectrum is consistent with
the
predicted structure.
O /~
-~ , ~/
~ ~
(N,rH 'CF3 CNf- NHy
/ ~
~y H ~y H
H \ H \
48 49
N-(N'-(4-benzoylphenylalanine) aminoethyl) cyclopamine (49). Aqueous
ammonia (0.5 mL of a 29% (w/w) solution in water, 7.6 mmoles) was added to a
solution of 48 (20.4 mg, 25.4 moles) in methanol (1 mL). The reaction was
stirred at
room temperature for 19 h and then evaporated to dryness by a stream of
nitrogen gas.
Purification by flash chromatography (Si02, step-wise gradient from 40:1:0.1
to 40:2:0.1
chloroform/methanol/triethylamine) yielded the amine as a colorless oil (17.9
mg, 25.4

CA 02386190 2002-04-11
WO 01/27135 PCT/US00/28479
- 124 -
moles, quant.). LRMS: not performed. 1H NMR: spectrum is consistent with the
predicted structure.
/~ , /~
, , .
~/ ~/
~
" H
NHZ J--f
/ ~1-1
p Fi ~ H
H ~
H
49 50
N-(N'-(N"-Levulinoyl (4-benzoylphenylalaninoyl)) aminoethyl) cyclopamine
(50). Triethylamine (5.30 L, 38.0 moles) and 47 (8.10 mg, 38.0 moles) were
added
to a solution of 49 (13.4 mg, 19.0 moles) in dichloromethane (0.5 mL). The
reaction
was stirred at room temperature for 4 h and then evaporated to dryness by a
stream of
nitrogen gas. Purification by flash chromatography (Si02, step-wise gradient
from
40:1:0 to 40:2:0.1 chloroform/methanol/triethylamine) yielded the diketone as
a
colorless oil (11.3 mg, 14.1 moles, quant.). LRMS: (ES+) calcd for C50H65N306
(M +
H): 804; found: 804. 1H NMR: spectrum is consistent with the predicted
structure.
~ ~
\ /
O
~",,rfex~, ~ ~`~'~
0
/ j-I / Fi
~ H ~ H
11
H \
50 51
3-Keto N-(N'-(N"-Levulinoyl (4-benzoylphenylalaninoyl)) aminoethyl)
cyclopamine (51). Dimethylsulfoxide (12.9 L, 182 moles) was added to a
solution of
oxalyl chloride (7.96 L, 91.2 moles) in dichloromethane (250 L) at -78 C.
After the
mixture was stirred at -78 C for 10 min, a solution of 50 (5.65 mg, 7.03
moles) in

CA 02386190 2002-04-11
WO 01/27135 PCT/US00/28479
-125-
dichloromethane (250 L) was added, and the reaction was stirred at -78 C for
another
30 min. The oxidation was completed by the addition of triethylamine (38.1 L,
273
moles) to the solution, which was stirred at -78 C for 10 min and then
allowed to
warm to room temperature. The reaction was quenched by the addition of
saturated
aqueous NaHCO3 (2 mL) was extracted with chloroform (2 x 5 mL). The resultant
organic layer was then dried over Na2SO4, and concentrated in vacuo.
Purification by
flash chromatography (Si02, step-wise gradient from 4:1 to 1:1 hexane/acetone)
yielded
the ketone as a white solid (1.4 mg, 1.75 moles, 25%). LRMS: (ES+) calcd for
C50H63N306 (M + H): 802; found: 802. 1H NMR: spectrum is consistent with the
predicted structure.
H O
OH
d O H O
O
52
N,N'-(4-Benzoylbenzoyl) (tert-butoxycarbonyl) lysine (52). 4-Benzoylbenzoic
acid N-hydroxysuccinimide ester (50.0 mg, 147 moles) was added to a solution
of tert-
butoxycarbonyl lysine (43.4 mg, 176 moles) in DMF (0.5 mL). 1 N NaOH (176 L,
176 moles) and water (74 L) was added to the reaction, and the mixture was
stirred
for 4 h at room temperature. 1 N NaOH was added again (352 L, 352 moles) and
the
solution was stirred overnight. The reaction mixture was then mixed with water
(2 mL)
and washed with diethyl ether (2 x 2 mL). The aqueous layer was acidified with
1 N HCl
until a pH of 2 was obtained, and the solution was extracted with ethyl
acetate (2 x 2
mL). The resultant organic layer was dried over MgSO4 and concentrated in
vacuo.
Purification by flash chromatography (Si02, step-wise gradient from 20:1:0.2
to 20:2:0.2
chloroform/methanol/acetic acid) yielded the benzophenone as colorless oil
(40.0 mg,
88.0 moles, 60%). LRMS: (ES+) calcd for C25H30N206 (M + H): 455; found: 455.
1H
NMR: spectrum is consistent with the predicted structure.

CA 02386190 2002-04-11
WO 01/27135 PCT/US00/28479
- 126 -
F3
H O H O
OH H
H O H
52 53
N,N'-(4-Benzoylbenzoyl) (trifluoroacetyl) lysine (53). Benzophenone 52 (3.55
mg, 78.1 moles) was dissolved in trifluoroacetic acid (0.5 mL, 6.49 mmoles),
and the
solution was stirred at room temperature for 1 h. The trifluoroacetic acid was
then
removed with a stream of nitrogen gas, and the residue was resuspended in
methanol
(0.5 mL). After triethylamine (134.8 L, 968 moles) and methyl
trifluoroacetate (24.3
L, 242 moles) were added to the methanol solution, the reaction was stirred
for 26 h
at room temperature. The reaction was evaporated to dryness in vacuo and
purification
by flash chromatography (Si02, step-wise gradient from 20:1:0.1 to 20:2:0.1
chloroform/methanoUacetic acid) yielded the trifluoroacetamide as a colorless
oil
(31.3 mg, 69.5 moles, 89%). LRMS: (ES+) calcd for C22H21N205F3 (M + H): 451;
found: 451. 1H NMR: spectrum is consistent with the predicted structure.
H 9
H
H
~/ C r H H
53 54
N,N'-(4-Benzoylbenzoyl) (trifluoroacetyl) lysine N-hydroxysuccinimide ester
(54). Disuccinimidyl carbonate (16.0 mg, 62.6 moles) was added to a solution
of 53
(28.2 mg, 62.6 moles) and pyridine (10.1 L, 125 moles) in acetonitrile (1.0
mL). The
reaction mixture was stirred at room temperature for 3 h, during which the
solution
became clear and evolved gas. The solution was added to ethyl acetate (10 mL),
washed
with 1 N HC1 (1 x 2 mL) and saturated aqueous NaHCO3 (1 x 2 mL), dried over
MgSO4, and concentrated in vacuo to yield a colorless oil (32.8 mg, 59.9
moles, 96%).

CA 02386190 2002-04-11
WO 01/27135 PCT/US00/28479
- 127 -
LRMS: (ES+) calcd for C26H24N307F3 (M + H): 548; found: 548. 1H NMR: spectrum
is consistent with the predicted structure
F3
H O
H
-fJHy
/` O H
O
/ ,~..~
~ FI H H
H \ H
17 55
N-(N'-(N",N"'-(4-Benzoylbenzoyl) (trifluoroacetyl) lysine) aminoethyl)
cyclopamine (55). Triethylamine (8.36 L, 60.0 moles) was added to a solution
of 54
(16.4 mg, 30.0 moles) in dichloromethane (0.5 mL). The solution became bright
yellow, indicating racemization of the amino acid a-carbon. A solution of 17
(13.6 mg,
30.0 moles) in dichloromethane (250 L) was then added to the yellow
solution. The
reaction mixture was stirred for 1 h at room temperature, during which it
became
colorless, and purification by flash chromatography (Si02, step-wise gradient
from 2:1
to 1:2 hexane/acaetone) yielded the cyclopamine derivative as a colorless oil
(15.8 mg,
17.8 moles, 59%, mixture of diastereomers). LRMS: (ES+) calcd for
C51H65N406F3
(M + H): 887; found: 887. 1H NMR: spectrum is consistent with the predicted
structure
F3
H O HZ
H H
H H Ao~O
/ ~H / 'H
F N ~ H
H H
55 56

CA 02386190 2002-04-11
WO 01/27135 PCT/US00/28479
- 128 -
N-(N'-(4-Benzoylbenzoyl) lysine) aminoethyl) cyclopamine (56). Compound
55 (13.0 mg, 14.7 moles) was dissolved in a 2 M solution of ammonia in
methanol (1.0
mL, 2.00 mmoles). The reaction was stirred at room temperature for 23 h and
then
evaporated to dryness by a stream of nitrogen gas. Purification by flash
chromatrography
(SiO2, step-wise gradient from 20:1:0.1 to 20:2:0.1
chloroform/methanoUtriethylamine)
yielded the amine as a white waxy solid (11.0 mg, 13.9 moles, mixture of
diastereomers, 95%). LRMS: (ES+) calcd for C49H66N405 (M + H): 791; found:
791.
1H NMR: spectrum is consistent with the predicted structure
H X
fi H
Hy
H
H
~ H I / ~ I
O H O
~Hl
H
H ~ H C H
56
H
H 57
N-(N'-(N",N"'-(4-Benzoylbenzoyl) (NI'll-digoxigenin 3-0-methylcarbonyl
aminocaproyl) lysine) aminoethyl) cyclopamine (57). Digoxigenin 3-0-
methylcarbonyl aminocaproic acid N-hydroxysuccinimide ester (5.0 mg, 7.59
moles)
and triethylamine (1.59 L, 11.4 moles) were added to a solution of 56 (4.5
mg, 5.69
moles) in dichloromethane (0.5 mL). After the solution was stirred for 2 h at
room
temperature, ethylenediamine (10 L, 150 moles) was added and the reaction
was
stirred for an addition 15 min at room temperature. Saturated aqueous NaHCO3
(2 mL)
was added to the mixture, which was then extracted with chloroform (5 mL, then
2 mL).
The organic layers were combined and dried over Na2SO4, and concentratedin
vacuo.

CA 02386190 2002-04-11
WO 01/27135 PCT/US00/28479
- 129 -
Purification by flash chromatography (Si02, step-wise gradient from 20:1 to
5:1
dichloromethane/methanol) yielded the digoxigenin derivative as white waxy
solid (3.5
mg, 2.62 moles, 46%). LRMS: (ES+) calcd for C80H11 tN5012 (M + H): 1337;
found:
1337. 1H NMR: spectrum is consistent with the predicted structure.
H H
Ff,,.
Hy
H
H
H
H O
~ }I
H
H ~ H H
56
~y H
H
58
N-(N'-(N",N"'-(4-Benzoylbenzoyl) (N""-biotinoyl aminocaproyl) lysine)
aminoethyl) cyclopamine (58). N-Biotinoyl aminocaproic acid N-
hydroxysuccinimide
ester (2.6 mg, 5.69 moles) and triethylamine (1.59 L, 11.4 moles) were
added to a
solution of 56 (4.5 mg, 5.69 moles) in DMF (250 L). After the reaction was
stirred for
2 h at room temperature, ethylenediamine (10 L, 150 moles) was added to the
solution, which was stirred for an addition 15 min at room temperature.
Saturated
aqueous NaHCO3 (2 mL) was added to the mixture, which was then extracted with
chloroform (1 x 2 mL). The organic layers were combined and dried over Na2SO4,
and
concentratedin vacuo. Purification by flash chromatography (Si02, step-wise
gradient
from 20:1 to 5:1 dichloromethane/methanol) yielded the digoxigenin derivative
as white
waxy solid (3.4 mg, 3.01 moles, 53%). LRMS: (ES+) calcd for C65H9tN708S (M +
H): 1130; found: 1130. 1 H NMR: spectrum is consistent with the predicted
structure.

CA 02386190 2002-04-11
WO 01/27135 PCT/US00/28479
-130-
~I I~ ~I I~
H ^ ^
C~ N~ ~ v vH
F3 H F3 H
O
46 59
N-(N'-Trifluoroacetyl (4-benzoylphenylalanine)) aminocaproic acid (59). A
solution of aminocaproic acid (10.8 mg, 80.5 moles) in water (100 L) and a
solution
of 46 (24.4 mg, 53.7 moles) in DMF (100 L) were combined and stirred for 45
min at
room temperature. The reaction was then acidified with 1 N HCl until a pH of 2
was
obtained, added to ethyl acetate (1 mL), washed with 1 N HCl (2 x 0.5 mL),
dried over
MgSO4, and evaporated to dryness by a stream of nitrogen gas yield a colorless
oil (25.9
mg, 54.1 moles, quant.). LRMS: not performed. 1H NMR: spectrum is consistent
with
the predicted structure.
FaC - H~~H F3AH H
H O
O O
59 60
N-(N'-Trifluoroacetyl (4-benzoylphenylalanine)) aminocaproic acid N-
hydroxysuccinimide ester (60). Disuccinimidyl carbonate (18.7 mg, 73.0 moles)
was
added to a solution of 59 (23.3 mg, 48.7 moles) and pyridine (7.88 L, 97.4
moles) in
acetonitrile (200 L). The reaction mixture was stirred at room temperature
for 12 h,
during which the solution became clear and evolved gas. The solution was added
to ethyl
acetate (1 mL), washed with 1 N HCl (1 x 0.5 mL) and saturated aqueous NaHCO3
(1 x
0.5 mL), dried over MgSO4, and evaporated to dryness by a stream of nitrogen
gas yield
a colorless oil (26.4 mg, 45.9 moles, 94%). LRMS: not performed. 1H NMR:
spectrum is consistent with the predicted structure.

CA 02386190 2002-04-11
WO 01/27135 PCT/US00/28479
- 131 -
H H
~HZ H 0
~N~ ~F3
/ \
\ /
~{ Fi ~ N
H \ H
17 61
N-(N'-(N"-(N"'-Tritluoroacetyl (4-benzoylphenylalaninoyl)) aminocaproyl)
aminoethyl) cyclopamine (61). Compound 60 (23.8 mg, 41.4 moles) and
triethylamine (11.5 L, 82.8 moles) were added a solution of 17 (15.7 mg,
34.5
moles) in dichloromethane (0.5 mL). The reaction was stirred for 1 h at room
temperature and purification by flash chromatography (Si02, step-wise gradient
from
2:1 to 1:2 hexane/acetone) yielded the benzophenone derivative as colorless
oil (5.3 mg,
5.79 moles, 17%). LRMS: not performed. 1H NMR: spectrum is consistent with
the
predicted structure.
H~~{ ^v ^v N-iCF3 H NHy
H ~O~ H
O / \ / \
N ~y H
F,
H H
61 62
N-(N'-(N"-(4-Benzoylphenylalaninoyl) aminocaproyl) aminoethyl)
cyclopamine (62). Aqueous ammonia (250 L of a 29% (w/w) solution in water,
3.80 mmoles) was added to a solution of 61 (5.3 mg, 5.79 moles) in methanol
(200
L). The reaction was stirred at room temperature for 18 h and then evaporated
to
dryness by a stream of nitrogen gas. Purification by flash chromatrography
(Si02, step-
wise gradient from 20:1:0.05 to 20:2:0.05 chloroform/methanol/triethylamine)
yielded
the amine as a colorless oil (4.0 mg, 4.88 moles, 84%). LRMS: not performed
1H
NMR: spectrum is consistent with the predicted structure.

CA 02386190 2002-04-11
WO 01/27135 PCT/US00/28479
- 132 -
H
CN\^^^ NHZ
n0
R
62
H H wH NH
CN~A H N~H 11
/ \
/ ~ \ /
0
~i H
H \ 63
N-(N'-(N"-(N"'-(N""-Biotinoyl aminocaproyl) (4-benzoylphenylalaninoyl))
aminocaproyl) aminoethyl) cyclopamine (63). N-Biotinoyl aminocaproic acid N-
hydroxysuccinimide ester (2.7 mg, 5.86 moles) and triethylamine (1.4 L, 9.76
moles) were added to a solution of 62 (4.0 mg, 4.88 moles) in DMF (250 L).
After
the reaction was stirred for 15 min at room temperature, it was added to
chloroform (2
mL), washed with saturated aqueous NaHCO3, dried over Na2SO4, and concentrated
in
vacuo. Purification by flash chromatography (Si02, step-wise gradient from
20:1:0.05 to
20:4:0.05 chloroform/methanol/triethylamine) yielded the biotin derivative as
white
solid (4.5 mg, 3.88 moles, 80%). LRMS: (ES+) calcd for C67H95N708S (M + H):
1158; found: 1158. 1H NMR: spectrum is consistent with the predicted
structure.
Example 3: In vivo testing
Methods: Female nude mice were injected subcutaneously with 5 million P2A6
fibrosarcoma cells derived from Ptc-/- embryonic fibroblasts. Three weeks
after
injection, each mouse had developed a discrete subcutaneous tumor. Each tumor
was
measured and treatments were initiated on the same day. Mice were treated once
daily
with intraperitoneal injections of tomatidine, cyclopamine, or KAAD-
cyclopamine 33
(one mouse per treatment) for four days. The mice were killed, and tumors were
measured and dissected out for histopathologic analysis. Tumor volumes were
calculated
as the product of length x width. The samples were paraformaldehyde-fixed and
paraffin-embedded, and slides were cut for hematoxylin and eosin staining and
for

CA 02386190 2006-08-30
-133- -
immunohistochemistry with polyclonal antibodies against the proliferation
marker Ki-67
*
(Novacastra NCL-Ki67p; assay performed according to manufacturer's
directions).
Results: As depicted in the graph, control treatment with tomatidine resulted
in
17% tumor growth over the treatment period whereas treatment with cyclopamine
decreased tumor size by 0.14% and treatment with KAAD-cyclopamine 33 decreased
tumor size by 19%.
To confirm these results, Ki-67 proliferation rates were determined by a
pathologist who was blinded to the treatment conditions. The magenta bars in
the graph
show proliferation rates for the three treatments. The proliferation rates for
tomatidine,
cyclopamine, and KAAD cyclopamine-treated tumors were 30%, 16%, and 12%
respectively.
Since the differentiation of many tumors correlates with prognosis, the
differentiation status of the tumors was examined. All tumors showed zonal
variation in
differentiation as illustrated in the photomicrographs of Figure 7. Compared
to more
poorly differentiated tumors, well differentiated fibrosarcomas have smaller,
less
crowded nuclei separated by relatively abundant pink collagen. As illustrated,
KAAD-
cyclopamine and cyclopamine (not shown) treated tumors showed a greater degree
of
differentiation than did the tomatidine-treated control.
Equivalents
Those skilled in the art will recognize, or be able to ascertain using no more
than
routine experimentation, many equivalents to the specific embodiments of the
invention
described herein. Such equivalents are intended to be encompassed by the
following
claims.
*Trade-mark

Representative Drawing

Sorry, the representative drawing for patent document number 2386190 was not found.

Administrative Status

2024-08-01:As part of the Next Generation Patents (NGP) transition, the Canadian Patents Database (CPD) now contains a more detailed Event History, which replicates the Event Log of our new back-office solution.

Please note that "Inactive:" events refers to events no longer in use in our new back-office solution.

For a clearer understanding of the status of the application/patent presented on this page, the site Disclaimer , as well as the definitions for Patent , Event History , Maintenance Fee  and Payment History  should be consulted.

Event History

Description Date
Inactive: Expired (new Act pat) 2020-10-13
Common Representative Appointed 2019-10-30
Common Representative Appointed 2019-10-30
Inactive: Late MF processed 2010-10-18
Letter Sent 2010-10-13
Grant by Issuance 2009-04-14
Inactive: Cover page published 2009-04-13
Inactive: Correspondence - Prosecution 2009-03-17
Letter Sent 2009-02-10
Inactive: Office letter 2009-02-09
Inactive: Final fee received 2009-02-03
Pre-grant 2009-01-30
Inactive: Final fee received 2009-01-30
Notice of Allowance is Issued 2008-11-25
Letter Sent 2008-11-25
Notice of Allowance is Issued 2008-11-25
Inactive: IPC assigned 2008-11-24
Inactive: Approved for allowance (AFA) 2008-11-17
Amendment Received - Voluntary Amendment 2008-09-29
Inactive: S.30(2) Rules - Examiner requisition 2008-04-04
Amendment Received - Voluntary Amendment 2008-01-03
Inactive: S.30(2) Rules - Examiner requisition 2007-07-03
Amendment Received - Voluntary Amendment 2007-05-11
Inactive: S.30(2) Rules - Examiner requisition 2006-11-15
Amendment Received - Voluntary Amendment 2006-08-30
Inactive: S.30(2) Rules - Examiner requisition 2006-05-17
Letter Sent 2006-03-21
Change of Address Requirements Determined Compliant 2006-03-08
Correct Applicant Request Received 2006-02-08
Change of Address or Method of Correspondence Request Received 2006-02-08
Inactive: Single transfer 2006-02-08
Amendment Received - Voluntary Amendment 2004-11-05
Letter Sent 2004-05-17
All Requirements for Examination Determined Compliant 2004-05-04
Request for Examination Requirements Determined Compliant 2004-05-04
Request for Examination Received 2004-05-04
Letter Sent 2002-09-30
Inactive: Cover page published 2002-09-27
Inactive: Notice - National entry - No RFE 2002-09-25
Inactive: First IPC assigned 2002-09-25
Application Received - PCT 2002-06-21
Inactive: Single transfer 2002-05-15
National Entry Requirements Determined Compliant 2002-04-11
Application Published (Open to Public Inspection) 2001-04-19

Abandonment History

There is no abandonment history.

Maintenance Fee

The last payment was received on 2008-09-19

Note : If the full payment has not been received on or before the date indicated, a further fee may be required which may be one of the following

  • the reinstatement fee;
  • the late payment fee; or
  • additional fee to reverse deemed expiry.

Patent fees are adjusted on the 1st of January every year. The amounts above are the current amounts if received by December 31 of the current year.
Please refer to the CIPO Patent Fees web page to see all current fee amounts.

Owners on Record

Note: Records showing the ownership history in alphabetical order.

Current Owners on Record
JOHNS HOPKINS UNIVERSITY SCHOOL OF MEDICINE
Past Owners on Record
ANSSI J. TAIPALE
JAMES K. CHEN
PHILIP A. BEACHY
Past Owners that do not appear in the "Owners on Record" listing will appear in other documentation within the application.
Documents

To view selected files, please enter reCAPTCHA code :



To view images, click a link in the Document Description column. To download the documents, select one or more checkboxes in the first column and then click the "Download Selected in PDF format (Zip Archive)" or the "Download Selected as Single PDF" button.

List of published and non-published patent-specific documents on the CPD .

If you have any difficulty accessing content, you can call the Client Service Centre at 1-866-997-1936 or send them an e-mail at CIPO Client Service Centre.


Document
Description 
Date
(yyyy-mm-dd) 
Number of pages   Size of Image (KB) 
Description 2002-04-10 133 5,878
Drawings 2002-04-10 9 337
Abstract 2002-04-10 1 47
Claims 2002-04-10 21 577
Description 2006-08-29 133 5,907
Claims 2006-08-29 12 463
Claims 2007-05-10 6 232
Claims 2008-01-02 7 227
Claims 2008-09-28 7 232
Reminder of maintenance fee due 2002-09-24 1 110
Notice of National Entry 2002-09-24 1 192
Courtesy - Certificate of registration (related document(s)) 2002-09-29 1 112
Acknowledgement of Request for Examination 2004-05-16 1 176
Courtesy - Certificate of registration (related document(s)) 2006-03-20 1 105
Commissioner's Notice - Application Found Allowable 2008-11-24 1 163
Late Payment Acknowledgement 2010-10-17 1 164
Maintenance Fee Notice 2010-10-17 1 171
Late Payment Acknowledgement 2010-10-17 1 164
PCT 2002-04-10 2 92
PCT 2002-04-11 1 32
PCT 2002-04-11 13 655
Correspondence 2006-02-07 2 50
Correspondence 2009-02-08 1 15
Correspondence 2009-01-29 1 37
Correspondence 2009-02-02 1 38
Correspondence 2009-02-09 1 13
Correspondence 2009-02-17 2 71